WO2019055829A1 - Methods for detecting cancer biomarkers - Google Patents

Methods for detecting cancer biomarkers Download PDF

Info

Publication number
WO2019055829A1
WO2019055829A1 PCT/US2018/051150 US2018051150W WO2019055829A1 WO 2019055829 A1 WO2019055829 A1 WO 2019055829A1 US 2018051150 W US2018051150 W US 2018051150W WO 2019055829 A1 WO2019055829 A1 WO 2019055829A1
Authority
WO
WIPO (PCT)
Prior art keywords
mutation
hist1h3b
h3f3a
plasma
tumor
Prior art date
Application number
PCT/US2018/051150
Other languages
French (fr)
Inventor
Javad NAZARIAN
Eshini PANDITHARATNA
Original Assignee
Nazarian Javad
Panditharatna Eshini
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nazarian Javad, Panditharatna Eshini filed Critical Nazarian Javad
Publication of WO2019055829A1 publication Critical patent/WO2019055829A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • the disclosure relates generally to detecting mutations in cancers of patients receiving cancer therapy and more particularly to detecting genetic mutations in brain cancers, including pediatric brain cancers.
  • midline anatomical brain structures e.g. pons, thalamus, spinal cord
  • DIPG diffuse intrinsic pontine glioma
  • MLGs harbor mutations in genes encoding canonical histones H3.1 (HIST1H3B/C), H3.2
  • the disclosure provides new methods for diagnosing cancers, including pediatric brain tumors.
  • the disclosure provides a method of determining the efficacy of a cancer therapy, said method comprising:
  • step c) comprises performing a method selected from the group consisting of quantitative polymerase chain reaction (qPCR), quantitative real-time polymerase chain reaction (qRTPCR), digital droplet PCR, (ddPCR), sequencing, northern blotting, or Southern blotting.
  • qPCR quantitative polymerase chain reaction
  • qRTPCR quantitative real-time polymerase chain reaction
  • ddPCR digital droplet PCR
  • the disclosure provides a method of treating cancer in a subject in need thereof, said method comprising:
  • step a) comprises performing digital PCR or drop digital PCR (ddPCR).
  • the disclosure provides a method of detecting a mutation in one or more genes selected from the group consisting of H3F3A, HIST1H3B, HIST1H3C, HIST2H3C, ACVRl, PPMID, PIK3R1, PIK3CA, IDHl, and BRAF in a blood or plasma sample from a human subject between 1 and 18 years of age, said method comprising performing digital PCR or droplet digital PCR (ddPCR) on the blood or plasma sample to determine the presence of a mutation in one or more genes selected from the group consisting of H3F3A, HIST1H3B, HIST1H3C, HIST2H3Q ACVRl, PPMID, PIK3R1, PIK3CA, IDHl, and BRAF in a biological sample.
  • the subject has cancer.
  • the cancer is a brain tumor.
  • the brain tumor is a diffuse glioma or a diffuse intrinsic potine gliom
  • the biological sample is a fluid.
  • the biological fluid is blood, plasma, serum, CSF, or urine.
  • the cancer is a brain tumor. In one embodiment, the cancer is a pediatric brain tumor. In some embodiments, the brain tumor is a diffuse glioma or a diffuse intrinsic pontine glioma (DIPG).
  • DIPG diffuse intrinsic pontine glioma
  • one or more genes comprise H3F3A.
  • the mutation is a K27M mutation in H3F3A.
  • the method further comprises determining the presence of a mutation in at least one other gene selected from the group consisting of HIST1H3B, HIST1H3C, HIST2H3C, ACVRl, PPMID, PIK3R1, PIK3CA, IDHl, and BRAF.
  • the mutation is a K27M mutation in H3F3A.
  • the method further comprises determining the presence of a mutation in at least one other gene selected from the group consisting of HIST1H3B, ACVRl, PPMID, PIK3R1, and PIK3CA. In another aspect of this embodiment, of any of the above methods, the method further comprises determining the presence of a mutation in at least one other gene selected from the group consisting of HIST1H3B, ACVRl, PPMID, and PIK3R1.
  • the disclosure provides an oligonucleotide probe comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 19-36, wherein the probe further comprises a label.
  • the disclosure provides a composition comprising at least one oligonucleotide probe comprising one of SEQ ID NOs: 19-36, wherein the probe further comprises a label.
  • FIGS. 1A and IB provide an overview of biofluid specimens utilized for detection of ctDNA in diffuse midline glioma patients.
  • FIG. 1A provides demographic, clinical, and molecular information for patients analyzed herein. Genomic alterations were obtained by tumor sequencing data generated for the PNOC003 trial, biorepository sequencing project, or other clinical studies.
  • FIG. IB is a flow chart of biofluid samples analyzed herein. Patients were either enrolled in PNOC003 trial (DIPG) or Children's National (CN) brain tumor biorepository (MLG); *patients enrolled on 1339 to donate specimens at postmortem.
  • DIPG PNOC003 trial
  • CN Children's National
  • MLG brain tumor biorepository
  • FIGS. 2A-N are graphical representations of genomic DNA validation for specific detection of mutant and wild type alleles of genes encoding driver mutations in pedatric diffuse midline gliomas. Shown are representative ddPCR plots for mutation detection using tumor genomic DNA from seven MLG patients and two non-CNS diseased brain tissue controls.
  • FIGS. 2A-2G are from tumor tissue;
  • FIGS. 2H-2N are from two non-CNS diseased brain tissue controls.
  • FIG. 3A-G are graphical representations of results obtained when assessing the sensitivity of a ddPCR platform with pre-amplification quantities of 2500 pg, 250 pg, 25 pg, and 2.5 pg of input DNA Genomic DNA.
  • the DNA was obtained from frozen tumor tissue obtained at postmortem, from a H3.3 K27M mutant DIPG patient was diluted and used to assess for sensitivity.
  • FIGS. 3A-3C are samples assessed without pre-amplification;
  • FIGS. 3D-3G are from samples assessed with preamplification.
  • FIGS. 4A-4B are graphical representations assessing the specificity of s ddPCR platform by testing for H3.3 K27M in H3 wildtype DIPG tumor tissue and liquid bio me.
  • FIG. 4A shows matched tumor tissue and CSF collected at postmortem from the same H3 WT DIPG patient who was negative for H3F3A p.K27M.
  • FIG. 4B shows that plasma collected at initial diagnosis was negative for H3F3A p.K27M in three H3 wild type DIPG patients.
  • FIG. 5A-B show schematics of biofluid specimen processing for ddPCR analysis.
  • FIG. 5A shows that genomic DNA was isolated from tumor tissue and used for ddPCR, while plasma and CSF were frozen upon collection, processed for ctDNA isolation, preamplified, and analyzed by ddPCR.
  • FIG. 5B shows that circulating DNA isolated from biofluid specimens were analyzed in a three-step method by ddPCR, which includes dropletization (by a'Source' machine), PCR, followed by detection in the 'Sense' machine.
  • An example is shown for H3F3A p.K27M detection, where LNA probes detect a single nucleotide change differentiating mutant and wild type alleles during PCR.
  • FIGS. 6A-F are graphic and pictorial representations showing detection of an histone 3 p.K27M mutation (H3K27M) ctDNA in biofluids of patients diagnosed with midline glioma.
  • FIG. 6A shows detection of H3K27M ctDNA in CSF collected throughout a disease.
  • FIG. 6B shows significantly higher ctDNA MAFs for H3K27M were detected in CSF compared to plasma.
  • FIG. 6C shows higher H3K27M MAFs were present in CSF compared to plasma in the same patient.
  • FIG. 6D shows that plasma ctDNA collected at diagnosis in DIPG patients enrolled in the PNOC003 clinical trial analyzed for H3K27M.
  • FIG. 6E shows that detection of H3K27M plasma ctDNA at post radiation in two patients that lacked detection at diagnosis.
  • FIG. 6F shows an Increase in tumor volume by MRI at post radiation compared to diagnosis in patients shown in FIG. 6C.
  • FIGS. 7A-F show multiplexed detection of oncohistone and partner mutations in tumor and CSF of MLG patients. Genomic DNA and CSF ctDNA were analyzed from the same MLG patient for multiplexed detection of mutations.
  • FIGS. 8A-C show CSF collected from various neuroanatomical locations in MLGs. Bar graphs represent average for H3K27M MAF detection from technical duplicates, and error bars represent standard error of mean.
  • FIG. 8A shows ignificantly higher H3K27M MAF was detected in CSF collected from adjacent sites to tumor compared to distant in MLG patients.
  • FIG. 8B shows Cohort based analysis of CSF collected from DIPG patients from different neuroanatomical locations, indicating higher detection in locations closer to brainstem.
  • FIG. 8C shows matched analysis for CSF collected from lateral ventricles and lumbar spine at postmortem from the same DIPG patient, showing similar trends as in (FIG. 8A) and (FIG. 8B).
  • FIGS. 9A-D show detection of a novel histone 3 mutation in fluid present in a brainstem tumor cyst found in a DIPG patient at postmortem.
  • FIG. 9A is an image of a cyst (indicated by an arrow) in fresh tissue at postmortem.
  • FIG. 9B is an MRI image captured a month before postmortem indicating cyst in pontine tumor.
  • FIGS. 9C-9D show that higher MAF levels were found in cyst fluid (41%) compared to tumor tissue (34%), and CSF (38%) for H3F3A p.K27M.
  • FIGS 10A-B shows detection of H3.3 K27M ctDNA in CSF at postmortem in a DIPG patient that tested negative in CSF collected at diagnosis.
  • FIG. 10A shows detection of
  • FIG.10B shows the results of an MRI imaging at diagnosis and postmortem for the same patient CSF was analyzed for in FIG. 10A.
  • FIGS. 11A-F show that a temporal analysis of plasma ctDNA matches response to therapy in DIPG patients.
  • FIG. 11 A shows serial plasma ctDNA analysis of changes in H3K27M throughout course of treatment in patients that followed PNOC003 recommended therapy.
  • FIG. 1 IB shows dynamic changes in plasma ctDNA and MRI tumor measurements in one patient in response to therapy. FLAIR and Tl-weighted post- gadolinium MRI images prior to treatment demonstrating a prominent expansile pontine mass and an additional focus within right cerebellar hemisphere that demonstrates evidence of enhancement. After treatment, the pontine and right cerebellar hemisphere lesions decreased in size.
  • FIG. 11C shows fluctuations in plasma ctDNA and MRI tumor volumes during therapy.
  • FIG. 1 ID shows a significant decrease in plasma ctDNA and MRI tumor volumes in response to radiation therapy.
  • FIG. 1 IE shows a decrease in plasma ctDNA and MRI tumor burden from diagnosis to pre-cycle 3 of PNOC003 recommended
  • FIGS. 12A-12N show longitudinal changes in plasma ctDNA in association with MR imaging findings and clinical assessments.
  • Each line graph represents plasma ctDNA and MRI changes in an individual patient diagnosed with DIPG.
  • the red line depicts MRI tumor measurements and black line depicts changes in temporal plasma ctDNA. Error bars are standard error of mean for technical triplicates of plasma ctDNA assessed for MAF of H3K27M.
  • the colored legend at the bottom of each figure indicates the time point of plasma ctDNA and MRI assessment during course of disease: green for initial diagnosis/biopsy, grey for during therapy, red for tumor growth, and black for the end of therapy following tumor growth/progression.
  • FIG. 13A-C are histograms showing bio fluid ctDNA and tumor spread as assessed by MRI, genomic, and/or histological studies.
  • FIG. 13A shows tumor extension beyond site of primary tumor (pons or thalamus) as determined based on MRI obtained prior to postmortem, or molecular and/or histopathology of autopsied whole brain specimens.
  • CSF ctDNA was higher in 18 MLG patients with tumor extension as compared to three MLG patients without tumor spread. Tumor involvement beyond pons was assessed by MRI review of patients enrolled in PNOC003.
  • Plasma ctDNA and MRI collected from DIPG patients were analyzed (FIG. 13B) at initial diagnosis and (FIG. 13C) at time points during the course of disease (initial diagnosis, during therapy, and tumor growth). Unlike in CSF, ctDNA levels in plasma were not higher in DIPG patients with tumor spread.
  • circulating tumor DNA to monitor disease progression is non/minimally- invasive method that has been increasingly employed for disease monitoring in adult cancers including glioblastoma (GBM), melanoma, lung, breast, and colon cancersi3-n74 ,however such studies have not been applied to the pediatric population.
  • GBM glioblastoma
  • melanoma melanoma
  • lung breast
  • colon cancersi3-n74 colon cancersi3-n74
  • Described herein is a sensitive, specific, rapid and minimally-invasive method for tumor surveillance of pediatric MLGs by monitoring the liquid biome.
  • plasma collected through an ongoing clinical trial (PNOC003; NCT 2274987) conducted by the Pacific Pediatric Neuro-Oncology Consortium (PNOC)
  • PNOC003; NCT 2274987 conducted by the Pacific Pediatric Neuro-Oncology Consortium
  • MAF mutation allelic frequency
  • the disclosure provides a robust application of the use of ctDNA for tumor profiling, and assessment of tumor response to therapy in pediatric patients with MLGs. These results show the feasibility of incorporating liquid biopsy as a sensitive and minimally invasive tool to inform clinical management for children with MLGs
  • circulating tumor DNA to monitor disease progression is a noninvasive method that has been shown to be successful in adult glioblastoma, lung cancer, melanoma, breast cancer, and colon cancer (Tie et al. (2016) Sci. Transl. Med., 8(346):346ra92; Tsao et al. (2015) Sci. Rep., 5:11198, Garcia-Murillas et al. (2015) Sci Transl Med.,
  • ctDNA is detectable in less than 50% of primary brain tumors (Bettegowda et al. (2014) Sci Transl. Med., 6(224):224ra24) and that allelic frequency of H3F3A is low (Lewis et al. (2013) Science 340(6134): 857-61 ; Bender et al. (2013) Cancer Cell
  • ctDNA was molecularly characterized in cerebrospinal fluid (CSF) and plasma, and the clinical utility of ctDNA to monitor tumor burden in pediatric midline gliomas (MLGs) was assessed.
  • CSF cerebrospinal fluid
  • MLGs pediatric midline gliomas
  • methods are performed using highly sensitive PCR-based sequencing method, such as digital droplet PCR.
  • digital PGR refers to PCR wherein the sample is divided into discrete subunits prior to amplification by PGR.
  • the sample may be separated into thousands or millions of partitions, each containing either zero or one (or, at most, a few) template molecules.
  • Fluorescent probes may be used to identify amplified target DNA in the partitions.
  • Samples containing amplified product e.g., fluorescent
  • those without product e.g., no fluorescence
  • the ratio of positives to negatives in each sample can be used for quantification. For example, Poisson statistics can be used to determine the absolute template quantity without the need to consider the number of amplification cycles.
  • ddPCR droplet digital PCR
  • the PCR solution is divided into smaller reactions (e.g., picoliter sized) through a water oil emulsion technique, which are then made to ran PCR individually.
  • the PCR sample is partitioned into nanoliter-size samples and encapsulated in oil droplets, (see, e.g., Hinson et al. (2011) Anal. Chem., 83:8604-8610; Pinheiro et al. (2012) Anal. Chem.,
  • PCR process is well known in the art and includes, for example, methods described in US Patent No. 9,984,201, the contents of which are incorporated by reference herein in their entirety. These methods include, e.g., reverse transcription PCR, ligation mediated PCR, digital PCR (dPCR), or droplet digital PCR (ddPCR).
  • dPCR digital PCR
  • ddPCR droplet digital PCR
  • PCR is carried out as an automated process with a thermostable enzyme.
  • the temperature of the reaction mixture is cycled through a denaturing region, a primer annealing region, and an extension reaction region automatically.
  • Machines specifically adapted for this purpose are commercially available.
  • amplified sequences are also measured using invasive cleavage reactions such as the InvaderTM technology (Zou et al, 2010, Association of Clinical Chemistry (AACC) poster presentation on Jul. 28, 2010, "Sensitive Quantification of Methylated Markers with a Novel Methylation Specific Technology; and U.S. Pat. No. 7,011,944 (Prudent et al)).
  • InvaderTM technology Zaou et al, 2010, Association of Clinical Chemistry (AACC) poster presentation on Jul. 28, 2010, "Sensitive Quantification of Methylated Markers with a Novel Methylation Specific Technology; and U.S. Pat. No. 7,011,944 (Prudent et al)).
  • next generation sequencing technologies are widely available. Examples include the 454 Life Sciences platform (Roche, Branford, Conn.) (Margulies et al. 2005 Nature, 437, 376-380); Ulumina's Genome Analyzer, GoldenGate Methylation Assay, or Infinium
  • Methylation Assays i.e., Infinium HumanMethylation 27K BeadArray or VeraCode GoldenGate methylation array (Illumina, San Diego, Calif.; Bibkova et al, 2006, Genome Res. 16, 383-393; U.S. Pat. Nos. 6,306,597 and 7,598,035 (Macevicz); U.S. Pat. No. 7,232,656 (Balasubramanian et al.)); QX200.TM. Droplet DigitaLTM. PCR System from Bio-Rad; or DNA Sequencing by Ligation, SOLiD System (Applied Biosystems/Life Technologies; U.S. Pat. Nos. 6,797,470,
  • methods are performed using digital droplet PCR (ddPCR).
  • ddPCR digital droplet PCR
  • digital droplet PCR is one of the most sensitive methods compared to Sanger sequencing, quantitative PCR, and next generation sequencing techniques (Diaz et al. (2014) J. Clin. Oncol. 32(6):579-86; Baker, M. (2012) Nat Meth., 9:541-544).
  • the nucleic acids may be quantified by counting the sub- samples that contain PCR end-product (positive reactions) and the sub- samples containing no PCR end-product (negative reactions) taking into account the Poisson distribution.
  • Digital PCR dPCR
  • dPCR Digital PCR
  • Digital droplet PCR as used herein relates to a digital PCR method in which the initial sample is sub-divided into several droplets constituting the sub- samples.
  • ddPCR was used to detect major mutations in ctDNA of liquid biome specimens and to assess dynamic changes in ctDNA during a disease course.
  • H3 27M partners with at least one additional driver mutation in cell cycle regulatory or growth factor signaling pathways (Nikbakht et al. (2016) Nat. Commun., 7:11185).
  • the presence of H3K27M and partner mutations in ctDNA was identified from liquid biome of MLGs.
  • the amount of detectable ctDNA in CSF and plasma was assessed with regard to clinical variables such as response to therapy, MRI tumor measurements, stage of disease, tumor spread, overall survival, and neuroanatomieal location of biofluid collection.
  • CSF samples were collected at various stages of disease (upfront, recurrence, and postmortem) from pediatric high grade midline gliomas.
  • DIPG diffuse intrinsic pontine glioma
  • a liquid biopsy assay using a sensitive and robust digital droplet PGR system on CSF and plasma samples was developed.
  • Major DIPG driver mutations including H3F3A (e.g., p. 27M),
  • HIST1H3B (e.g., p. 27M), ACVR1 (e.g., p.G328V/p.R206H), PPM ID (e.g., p.E525X),
  • PI 3R1 e.g., p.K567E
  • PI 3CA e.g., p.H1047R
  • Samples can also be analyzed for IDH1 (e.g., p,R132H) and BRAF (e.g., p.V600E) mutations.
  • IDH1 e.g., p,R132H
  • BRAF e.g., p.V600E
  • H3K27M was detected in ctDNA of plasma samples in xenograft models of DIPG.
  • biofluids from patients and animal models e.g., urine, blood, serum, plasma, CSF
  • a liquid biopsy can complement or in some cases eliminate the need for biopsies and inform recurrence and tumor response to treatment.
  • the method comprises determining the presence of a mutation in histone H3 (e.g., histone H3.3) and administering a therapy (e.g., administering at least one therapeutic agent to the subject) to the subject if the mutation in histone H3 is present.
  • the method may further comprise the step of obtaining a biological sample from the subject.
  • the biological sample of the methods may be a fluid or liquid such as blood, CSF, plasma, or urine.
  • the biological sample is blood or plasma.
  • the therapy administered to the subject is an epigenetic regulating drug such as, without limitation, SAHA and panobiiiostat.
  • the cancer detected and treated by the instant methods may be a brain tumor. More specifically, the cancer may be a pediatric brain tumor. In a particular embodiment, the cancer is a glioma, particularly a diffuse glioma or a diffuse intrinsic pontine glioma (DIPG).
  • the methods of the instant invention may comprise detenrdning the presence of a mutation(s) in circulating tumor DNA (ctDNA). In a particular embodiment, determining of the presence of the mutation(s) comprises perforating digital PCR. In a particular
  • the determining of the presence of the mutation comprises performing droplet digital PCR (ddPCR).
  • the digital PCR or ddPCR may be performed with at least one primer comprising any one of SEQ ID NOs: 1-18.
  • the digital PCR or ddPCR may be performed with primers comprising each of SEQ ID NOs: 1-18.
  • the digital PCR or ddPCR may be performed with at least one probe comprising any one of SEQ ID NOs: 19-36.
  • the digital PCR or ddPCR may be performed with probes comprising each of SEQ ID NOs: 19-36.
  • the instant methods comprise determining the presence of a mutation in histone H3 (e.g., histone H3.3).
  • the mutation in histone H3 is K27M.
  • the histone H3 is H3F3A.
  • the method comprises determining the presence of a mutation in H3F3A and at least one other gene (e.g., partner gene).
  • the other gene(s) may be, for example, selected from the group of HIST1H3B (e.g., p.K27M), HIST2H3C (e.g., p.K27M), ACVR1 (e.g., p.G328V/p.R206H), PPM1D (e.g., p.E525X), PIK3R1 (e.g., p.K567E), PIK3CA (e.g., p.H1047R), IDH1 (e.g., p.R132H; isocitrate dehydrogenase (NADP(+)) 1, cytosolic; Gene ID: 3417) and BRAF (e.g., p.V600E; B-Raf proto-oncogene, serine/threonine kinase
  • the other gene(s) is selected from the group of HIST1H3B (e.g., p.K27M), ACVR1 (e.g.,
  • p.G328V/p.R206H PPM1D (e.g., p.E525X), and PIK3R1 (e.g., p.K567E) (e.g., 1, 2, 3, or all 4 genes are analyzed). Sequence information for certain of these genes is provided below.
  • H3 histone family member 3A (H3F3A; SEQ ID NO: 37) Gene ID: 3020, Gen Bank Accession Nos. NM_002107.4 and NP_002098.1 (Note: the Met at position 1 is not included in amino acid numbering)
  • H3 family member b HIST1H3B; SEQ ID NO: 38
  • ACVRl Activin A receptor type 1 (ACVRl ; SEQ ID NO: 39) Gene ID: 90, Gen Bank Accession
  • Protein phosphatase, Mg2+/Mn2+ dependent ID (PPM1D; SEQ ID NO: 40) Gene ID: 84, Gen Bank Accession Nos. NM bend003620.3 and NPJ303611.1
  • Phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1 ; SEQ ID NO: 41) Gene ID: 529 Gen Bank Accession Nos. NM_181523.2 and NP_852664.1
  • Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA; SEQ ID NO: 42) Gene ID: 5290, Gen Bank Accession Nos. NM_006218.3 and NP_006209.2
  • the method comprises 1) administering a cancer therapy to a subject in need thereof, 2) obtaining a biological sample from the subject after administration of the cancer therapy to the subject, and 3) determining the presence of a mutation in in the biological sample (e.g., in histone H3 (e.g., histone H3.3)).
  • the method may comprise obtaining multiple biological samples over time from the subject after administration of the cancer therapy to the subject.
  • the method may comprise obtaining a biological sample from the subject before administration of the cancer therapy to the subject (e.g., a baseline).
  • the mutation e.g., in histone H3
  • the cancer therapy administered to the subject is effective (e.g., inhibiting tumor growth).
  • the biological sample of the methods may be a fluid or liquid such as blood, CSF, plasma, serum, or urine.
  • the biological sample is blood or plasma.
  • the cancer monitored by the instant methods may be a brain tumor. More specifically, the cancer may be a pediatric brain tumor. In a particular embodiment the cancer is a glioma, particularly a diffuse glioma or a diffuse intrinsic pontine glioma (DIPG).
  • DIPG diffuse intrinsic pontine glioma
  • the cancer therapy administered to the subject may comprise any type of therapy.
  • the cancer therapy may comprise the administration of at least one chemotherapeutie agent (e.g., a small molecule).
  • the cancer therapy may include radiation therapy.
  • the methods of the instant invention may comprise determining the presence of the mutation in circulating tumor DNA (ctDNA).
  • determining of the presence of the mutation comprises performing digital PGR.
  • the determining of the presence of the mutation comprises performing droplet digital PGR (ddPCR).
  • the digital PGR or ddPCR may be performed with at least one primer comprising any one of SEQ ID NOs: 1-14 or a portion thereof.
  • the digital PGR or ddPCR may be performed with at least one probe comprising any one of SEQ ID NOs: 19-36.
  • the digital PGR or ddPCR may be performed with probes comprising each of SEQ ID NOs: 19-36.
  • the instant methods comprise determining the presence of a mutation (e.g., in histone H3, such as histone H3.3).
  • the method comprises determining the presence of a mutation in a gene(s) selected from the group of H3F3 A (e.g., p.K27M), HIST1H3B (e.g., p.K27M), HIST2H3C (e.g., p.K27M), ACVR1 (e.g., p.G328V/p.R206H), PPM1 (e.g., p.E525X), FK3R1 (e.g., p.
  • H3F3 A e.g., p.K27M
  • HIST1H3B e.g., p.K27M
  • HIST2H3C e.g., p.K27M
  • ACVR1 e.g., p.G328V/p.R206H
  • PPM1 e.
  • PIK3CA e.g., p.H1047R
  • IDH1 e.g., p.R132H; isocitrate dehydrogenase (NADP(+)) 1, cytosolic (Gene ID:3417)
  • BRAF e.g., p.V600E; B-Raf proto-oncogene, serine/threonine kinase (Gene ID: 673)
  • the gene(s) is selected from the group of H3F3A (e.g., p.K27M), HIST1H3B (e.g., p.K27M), ACVR1 (e.g.,
  • the other gene(s) is selected from the group of HIST1H3B (e.g., p.K27M), ACVR1 (e.g.,
  • the mutation in histone H3 is K27M.
  • the histone H3 is H3F3A.
  • the method comprises determining the presence of a mutation in H3F3A and at least one other gene. A decrease in the presence of the mutations over time (e.g., when multiple biological samples are obtained after therapy) or compared to the amount observed prior to therapy, indicates that the cancer therapy administered to the subject is effective (e.g., inhibiting tumor growth).
  • oligonucleotide probes are also provided with the instant invention.
  • the probes are designed to have high affinity and specificity to the target site (e.g., the mutations set forth herein and, optionally, the wild- type gene).
  • oligonucleotide probe(s) target a gene (e.g., wild-type and/or mutant) selected from the group of H3F3A (e.g., p.K27M), HIST1H3B (e.g., p.K27M), HIST2H3C (e.g., p.K27M), ACVR1 (e.g., p.G328V/p.R206H), PPM1D (e.g., p.E525X), PIK3R1 (e.g., p.K567E), PIK3CA (e.g., p.H1047R), IDH1 (e.g., p.R132H) and BRAF (e
  • the probes do not have an absolute requirement on length. However, the probes will typically be from about 10 to about 250 nucleotides, about 10 to about 100, about 10 about 50 nucleotides, about 10 to about 40 nucleotides, about 10 to about 30 nucleotides, about 10 to about 25 nucleotides, or about 10 to about 20 nucleotides. In a particular embodiment, the probe is at least about 10 nucleotides in length. The probe may be at least 85%, at least 90%, at least 95%, at least 97%, or, more preferably, 100% complementary to the target sequence. In a particular embodiment, the oligonucleotide probe is designed such that the mutation is towards the middle of the sequence of the probe (e.g., within the middle third of the probe length).
  • the probe may comprise at least one nucleotide analog.
  • the nucleotide analogs may be used to increase annealing affinity and/or specificity and/or resistance to degradation.
  • LNA locked nucleic acid
  • Nucleotide analogs include, without limitation, nucleotides with phosphate modifications comprising one or more phosphorothioate, phosphorodithioate, phosphodiester, methyl phosphonate, phosphoramidate, methylphosphonate, phosphotriester, phosphoroaridate, morpholino, amidate carbamate, carboxymethyl, acetamidate, polyamide, sulfonate, sulfonamide, sulfamate, formacetal, thioformacetal, and/or alkylsilyl substitutions; nucleotides with modified sugars; and nucleotide mimetics such as, without limitation, peptide nucleic acids (PNA), morpholino nucleic acids, cyclohexenyl nucleic acids, anhydrohexitol nucleic acids, glycol nucleic acid, threose nucleic acid, and locked nucleic acids (LNA).
  • PNA peptide nucle
  • the probes comprise at least one locked nucleic acid.
  • the probes may comprise one of SEQ ID NOs: 19-36 or a sequence with at least 85%, at least 90%, at least 95%, or at least 97% identity to one of SEQ ID NOs: 19-36.
  • the probe comprises one of SEQ ID NOs: 19-36.
  • the probes may comprise additional nucleotides 5' or 3' to the included SEQ ID NO.
  • the probe may comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9,10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more nucleotides 5' or 3' to the included SEQ ID NO.
  • the additional sequences are complementary to the target sequence.
  • the probes of the instant invention may comprise one or more fluorescent probes / fluorophores and/or quenchers.
  • the fluorophores and or quenchers may be added to the 5' and or 3' termini of the probes.
  • the fluorophores and/or quenchers may also be added to internal part of the probes (e.g., a ZEN probe). Fluorophores and/or quenchers are well known in the art (see, e.g., IDT, Coralville, IA).
  • fluorophores and/or quenchers include, without limitation, 6-FAM, fluorescein, Cy3, Cy5, TAMRA, JOE, MAX, TET, Cy5.5, ROX, ATTO, TYE, Yakima Yellow®, HEX, TEX, Texas Red®, Iowa Black®, ZEN, and Alexa Fluor®.
  • the fluorophores and/or quenchers used allow for the determination of the presence of the wild- type and/or mutant allele in a sample at the same time (see, e.g., the Examples).
  • the fluorophores and/or quenchers create an energy transfer pair (e.g., fluorescence resonance energy transfer (FRET)) (e.g.,as set forth in Table 1).
  • FRET fluorescence resonance energy transfer
  • the probes comprise a fluorophore and/or quencher combination presented in Table 1.
  • the probe comprises any one of SEQ ID NOs: 19-36.
  • the probe comprises any one of SEQ ID NOs: 19-36 along with the modifications presented in Table 1.
  • compositions comprising at least one probe of the instant invention are also provided.
  • the composition is an aqueous solution.
  • the composition comprises at least one probe comprising any one of SEQ ID NOs: 19-36.
  • the composition comprises individual probes comprising each of SEQ ID NOs: 19-36.
  • compositions comprising at least one primer of the instant invention are also provided.
  • the composition is provided an aqueous solution.
  • the composition comprises at least one primer comprising any one of SEQ ID NOs: 1-18.
  • the composition comprises individual primers comprising each of SEQ ID NOs: 1-18.
  • “Pharmaceutically acceptable” indicates approval by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • a “carrier” refers to, for example, a diluent, adjuvant, preservative (e.g.,Thimersol, benzyl alcohol), anti-oxidant (e.g., ascorbic acid, sodium metabisulfite), solubilizer (e.g., polysorbate 80), emulsifier, buffer (e.g., Tris HC1, acetate, phosphate), antimicrobial, bulking substance (e.g., lactose, mannitol), excipient, auxiliary agent or vehicle (e.g., with which an active agent of the present invention is administered).
  • Pharmaceutically acceptable carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin.
  • Water or aqueous saline solutions and aqueous dextrose and glycerol solutions may be employed as carriers, particularly for injectable solutions.
  • Suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences” by E.W. Martin (Mack Publishing Co., Easton, PA); Gennaro, A. R., Remington: The Science and Practice of
  • small molecule refers to a substance or compound that has a relatively low molecular weight (e.g., less than 4,000, less than 2,000, particularly less than 1 kDa or 800 Da).
  • small molecules are organic, but are not proteins, polypeptides, or nucleic acids, though they may be amino acids or dipeptides.
  • treat refers to any type of treatment that imparts a benefit to a patient afflicted with a disease, including improvement in the condition of the patient (e.g., in one or more symptoms), delay in the progression of the condition, etc.
  • prevent refers to the prophylactic treatment of a subject who is at risk of developing a condition resulting in a decrease in the probability that the subject will develop the condition.
  • diagnosis refers to detecting and identifying a disease or disorder in a subject.
  • the term may also encompass assessing or evaluating the disease or disorder status (progression, regression, stabilization, response to treatment, etc.) in a patient known to have the disease or disorder.
  • the term "prognosis” refers to providing information regarding the impact of the presence of a disease or disorder (e.g., as determined by the diagnostic methods of the present invention) on a subject's future health (e.g., expected morbidity or mortality, the likelihood of getting or risk of cholestasis).
  • a disease or disorder e.g., as determined by the diagnostic methods of the present invention
  • future health e.g., expected morbidity or mortality, the likelihood of getting or risk of cholestasis.
  • prognosis refers to providing a prediction of the probable course and outcome of a disease/disorder and/or the likelihood of recovery from the disease/disorder.
  • the term "subject" refers to an animal, particularly a mammal, particularly a human.
  • composition refers to an amount effective to prevent, inhibit, treat, or lessen the
  • the treatment of a disease or disorder herein may refer to curing, relieving, and/or preventing the disease or disorder, the symptom(s) of it, or the predisposition towards it.
  • therapeutic agent refers to a chemical compound or biological molecule including, without limitation, nucleic acids, peptides, proteins, and antibodies that can be u ed to treat a condition, disease, or disorder or reduce the symptoms of the condition, disease, or disorder.
  • isolated refers to the separation of a compound from other components present during its production or from its natural environment. "Isolated” is not meant to exclude artificial or synthetic mixtures with other compounds or materials, or the presence of impurities that do not substantially interfere with the fundamental activity, and that may be present, for example, due to incomplete purification, or the addition of stabilizers.
  • a “biological sample” refers to a sample of biological material obtained from a subject, particularly a human subject, including a tissue, a tissue sample, cell(s), and a biological fluid (e.g., blood (e.g., whole blood), serum, plasma, urine, sweat, tears, saliva, mucosal secretions, sputum, CSF).
  • a biological fluid e.g., blood (e.g., whole blood), serum, plasma, urine, sweat, tears, saliva, mucosal secretions, sputum, CSF).
  • probe refers to an oligonucleotide, polynucleotide or nucleic acid, either RNA or DNA, which is capable of annealing with or specifically hybridizing to a nucleic acid with sequences complementary to the probe.
  • a probe may be either single- stranded or double- stranded. The exact length of the probe will depend upon many factors, including temperature, source of probe and use of the method.
  • the oligonucleotide probe typically contains about 10-250, about 10-100, about 10-50, about 15-30, about 15-25, or about 10-20 nucleotides.
  • the probes herein may be selected to be complementary to different strands of a particular target nucleic acid sequence. This means that the probes must be sufficiently complementary so as to be able to "specifically
  • the probe sequence need not reflect the exact complementary sequence of the target, although they may.
  • a non-complementary nucleotide fragment may be attached to the 5' or 3' end of the probe, with the remainder of the probe sequence being complementary to the target strand.
  • non- complementary bases or longer sequences can be interspersed into the probe, provided that the probe sequence has sufficient complementarity with the sequence of the target nucleic acid to anneal therewith specifically.
  • a probe may be tagged or labeled (i.e., attached to an entity making it possible to identify a compound to which it is associated (e.g., fluorescent or radioactive tag).
  • a label is selected from the group consisting of biotin, copper-DOTA, biotin-PEG3, aminooxyacetate, 19 FB, 18 FB, FITC-PEG 3 , fluorescein and fluorescein derivatives (e.g., 5-carboxy fluorescein).
  • the label is selected from the group consisting of 64 Cu DOT A, 68 Ga DOT A, 18 F, ⁇ C , 68 Ga, 89 Zr, i24 I, 86 Y, 94m Tc, n0ra Xn, "C and 76 Br.
  • primer refers to an oligonucleotide, either RN A or DNA, either single- stranded or double-stranded, which, when placed in the proper environment, is able to functionally act as an initiator of template-dependent nucleic acid synthesis.
  • suitable nucleoside triphosphate precursors of nucleic acids a polymerase enzyme, suitable cofactors and conditions such as appropriate temperature and H
  • the primer may be extended at its 3' terminus by the addition of nucleotides by the action of a polymerase or similar activity to yield a primer extension product.
  • the primer may vary in length depending on the particular conditions and
  • the oligonucleotide primer is typically about 10- 25 or more nucleotides in length, but can be significantly longer.
  • the primer must be of sufficient complementarity to the desired template to prime the synthesis of the desired extension product, that is, to be able to anneal with the desired template strand in a manner sufficient to provide the 3' hydroxyl moiety of the primer in appropriate juxtaposition for use in the initiation of synthesis by a polymerase or similar enzyme. It is not required that the primer sequence represent an exact complement of the desired template, though it may.
  • a non- complementary nucleotide sequence may be attached to the 5' end of an otherwise complementary primer.
  • non-complementary bases may be interspersed within the oligonucleotide primer sequence, provided that the primer sequence has sufficient complementarity with the sequence of the desired template strand to functionally provide a template-primer complex for the synthesis of the extension product.
  • oligonucleotide refers to sequences, primers and probes of the present invention, and is defined as a nucleic acid molecule comprised of two or more ribo- or deoxyribonucleotides, preferably more than three. The exact size of the oligonucleotide will depend on various factors and on the particular application and use of the oligonucleotide.
  • the respective fluorophores are added to the 5' end of the probes, and the quenchers are added to the middle and at the 3' end of the probe.
  • the symbol + following a nucleotide indicates a locked nucleic acid.
  • IABkFQ Iowa Black® FQ.
  • SEQ ID NO is provided in parentheses. The SEQ ID NOs represent the nucleotide sequences without the probes or modifications.
  • FIG. 1A This example illustrates a sensitive and specific platform for discriminating rare, low abundant, tumor-associated circulating DNA in pediatric patients with mid-line glioma (MLG) tumors.
  • MLG mid-line glioma
  • Fig. 1A Clinicopathological and genomic characteristics of the tumors of a pediatric MLG patient cohort are shown in Fig. 1A.
  • Patients were diagnosed with MLGs harboring various mutation combinations, including oncohistone variants: 94% harbored histone 3 mutations (79% with H3.3K27M, 15% with H3.1K27M), and 6% were H3 wild type.
  • the initial goal was to develop and validate a clinically relevant and minimally invasive Uquid biopsy platform suitable for detection and quantification of somatic mutations associated with pediatric MLGs.
  • Example 2 CSF and plasma harbor circulating tumor DNA indicative of driver mutations associated with pediatric MLGs.
  • Liquid biome specimens were analyzed from 84 subjects (48 MLG patients, and 36 non- CNS diseased controls), enrolled in an ongoing clinical trial PNOC003 (NCT 227498), and consented for the Children's National (CN) brain tumor biorepository (FIG. IB).
  • CSF samples were collected at a single time point through the CN biorepository at pre-treatment, during therapy, and at postmortem from 27 MLG patients, while serial sampling at pre-treatment and postmortem was available for one patient with DIPG.
  • Histone 3 mutant and wild type alleles were detected in 75% of CSF specimens collected at diagnosis, 67% of those collected during treatment, and 90% of those collected at postmortem (Fig. 2a).
  • H3 K27M- mutant ctDNA was detected in 89% of all CSF specimens analyzed from 27 MLGs, where 23 of these were confirmed to harbor oncohistones as assessed by tumor DNA analysis.
  • Example 3 Assessment of treatment response using ctDNA quantification.
  • Liquid biopsy is an emerging tool for diagnosing, and measuring efficacy of treatment in adult cancer patients. While molecular profiling of tumors is a localized method, a liquid biopsy approach provides a systemic molecular overview. ctDNA has been used to determine patient's mutation profiles, as a biomarker for molecular-based disease monitoring, and recurrence in adult chronic lymphocytic leukemia, breast, and colon cancer. 13 ' 16 ' 23 The only previously reported liquid biopsy approach for pediatric MLGs was established using Sanger sequencing.
  • the disclosure herein for the detection and quantification of tumor-associated circulating DNA using ddPCR allows for rapid, more sensitive, far less costly and less invasive method for surveying tumor mutations, and represents a key advance particularly for tumors with limited tissue acquisition or prohibitive sampling at multiple time points.
  • DIPG tumor cells disseminate throughout the brain during the course of disease.
  • Our ctDNA analysis in DIPG patients was indicative of tumor expansion beyond pons, where an increased amount of ctDNA in CSF was observed in patients who exhibited tumor spread.
  • Studies of a larger cohort in clinical settings are required to assess the statistical significance of our finding.
  • our results indicate the unique strength of liquid biopsy for assessing the molecular landscape of MLGs, and potential for longitudinal assessment of tumor response to therapy, which is a new tool that is complementary to MR imaging. Similar to the clinical utility of ctDNA for monitoring response to therapy with respect to MRIs in adult GBMs, despite a small sample size, we found significant reduction in ctDNA following RT.
  • Table 2 Assessing specificity of a ddPCR platform by testing non-CNS malignant pediatric CSF and plasma.
  • Average MAF values for non- CNS diseased plasma analyzed for H3F3A p.K27M mutation represent technical triplicates, all other average MAF values for plasma and CSF analyzed for H3F3A and HIST1H3B p.K27M represent technical duplicates.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Hospice & Palliative Care (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

Circulating tumor biomarkers and methods of use thereof are provided.

Description

METHODS FOR DETECTING CANCER BIOMARKERS
RELATED APPLICATIONS
This application claims priority to U.S. Provisional Application 62/559,049, entitled "Circulating Tumor Biomarkers and Methods of Use Thereof, filed September 15, 2017. The contents of the aforementioned application is hereby incorporated by reference herein in its entirety.
FIELD
The disclosure relates generally to detecting mutations in cancers of patients receiving cancer therapy and more particularly to detecting genetic mutations in brain cancers, including pediatric brain cancers.
BACKGROUND
Pediatric brain cancers are the leading cause of cancer related mortality in children under
14 years of age1. Among these, mahgnant tumors forming in midline anatomical brain structures (e.g. pons, thalamus, spinal cord) have dismal outcomes2. Children diagnosed with midline gliomas (MLGs) - including children with diffuse intrinsic pontine glioma (DIPG) - often die within a year of diagnosis.
MLGs harbor mutations in genes encoding canonical histones H3.1 (HIST1H3B/C), H3.2
{HIST2H3C), and non-canonical histone H3.3 (H3F3A) variants24. Thus, these tumors were recently re-classified by the World Health Organization (WHO) as diffuse midline glioma, H3 K27M-mutant5. While surgical biopsies for diagnosing brainstem tumors have proven safe, characterize tumor biology and potentially guide therapy6"7 the sensitive anatomical location of these tumors and the need for specialized expertise has prohibited a wider application of surgical biopsies, especially when performing biopsies for tumor recurrence. MR imaging combined with clinical examination is currently being used to assess therapy responses, both of which at times lack sensitivity and specificity.8"10 As new biologically targeted strategies, specifically immunotherapy approaches are entering the clinic for children with MLGs, the inability to accurately assess disease response, and treatment related molecular changes remain significant challenges.11"12 Despite ongoing biopsy- informed clinical trials, including convection-enhanced delivery (CED) of drugs, monitoring tumor response to treatment remains a challenge. Magnetic resonance imaging (MRI) is not a sensitive method for monitoring tumor burden and is often slow to reflect tumor progression and regression.
Therefore, there is a need for the development of safe and sensitive tests of cancer biomarkers for the brain.
SUMMARY
The disclosure provides new methods for diagnosing cancers, including pediatric brain tumors.
Accordingly, in one aspect, the disclosure provides a method of determining the efficacy of a cancer therapy, said method comprising:
a) administering a cancer therapy to a subject in need thereof;
b) obtaining a biological sample from the subject after administration of the cancer therapy to the subject; and
c) determining the presence of a mutation in one or more genes selected from the group consisting of H3F3A, HIST1H3B, HIST1H3C, HIST2H3C, ACVR1, PPM ID, PIK3R1, PIK3CA, IDH1, and BRAF in the biological sample,
wherein a decrease in the presence of the mutation(s) compared to the amount observed prior to administration of the cancer therapy indicates that the cancer therapy administered to the subject is effective. In one embodiment, steps b) and c) are repeated at least once, wherein a decrease in the presence of the mutation(s) over time indicates that the cancer therapy administered to the subject is effective. In some embodiments, step c) comprises performing a method selected from the group consisting of quantitative polymerase chain reaction (qPCR), quantitative real-time polymerase chain reaction (qRTPCR), digital droplet PCR, (ddPCR), sequencing, northern blotting, or Southern blotting.
In other aspects, the disclosure provides a method of treating cancer in a subject in need thereof, said method comprising:
a) determining the presence of a mutation in one or more genes selected from the group consisting of H3F3A, HIST1H3B, HIST1H3C, HIST2H3C, ACVR1, PPM ID, PIK3R1, PIK3CA, IDH1, and BRAF in a biological sample, and b) administering a cancer therapy to said subject if the mutation(s) is present. In some embodiments, step a) comprises performing digital PCR or drop digital PCR (ddPCR).
In other aspects, the disclosure provides a method of detecting a mutation in one or more genes selected from the group consisting of H3F3A, HIST1H3B, HIST1H3C, HIST2H3C, ACVRl, PPMID, PIK3R1, PIK3CA, IDHl, and BRAF in a blood or plasma sample from a human subject between 1 and 18 years of age, said method comprising performing digital PCR or droplet digital PCR (ddPCR) on the blood or plasma sample to determine the presence of a mutation in one or more genes selected from the group consisting of H3F3A, HIST1H3B, HIST1H3C, HIST2H3Q ACVRl, PPMID, PIK3R1, PIK3CA, IDHl, and BRAF in a biological sample. In some embodiments the subject has cancer. In one embodiment, the cancer is a brain tumor. In some embodiments, the brain tumor is a diffuse glioma or a diffuse intrinsic potine glioma (DIPG).
In some embodiments, the biological sample is a fluid. In some embodiments, the biological fluid is blood, plasma, serum, CSF, or urine.
In some embodiments, the cancer is a brain tumor. In one embodiment, the cancer is a pediatric brain tumor. In some embodiments, the brain tumor is a diffuse glioma or a diffuse intrinsic pontine glioma (DIPG).
In some embodiments, one or more genes comprise H3F3A. In one embodiment, the mutation is a K27M mutation in H3F3A. In another aspect of this embodiment, of any of the above methods, the method further comprises determining the presence of a mutation in at least one other gene selected from the group consisting of HIST1H3B, HIST1H3C, HIST2H3C, ACVRl, PPMID, PIK3R1, PIK3CA, IDHl, and BRAF. In another embodiment, the mutation is a K27M mutation in H3F3A. In another aspect of this embodiment, of any of the above methods, the method further comprises determining the presence of a mutation in at least one other gene selected from the group consisting of HIST1H3B, ACVRl, PPMID, PIK3R1, and PIK3CA. In another aspect of this embodiment, of any of the above methods, the method further comprises determining the presence of a mutation in at least one other gene selected from the group consisting of HIST1H3B, ACVRl, PPMID, and PIK3R1.
In other aspects, the disclosure provides an oligonucleotide probe comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 19-36, wherein the probe further comprises a label. In another aspect, the disclosure provides a composition comprising at least one oligonucleotide probe comprising one of SEQ ID NOs: 19-36, wherein the probe further comprises a label.
BRIEF DESCRIPTION OF THE FIGURES FIGS. 1A and IB provide an overview of biofluid specimens utilized for detection of ctDNA in diffuse midline glioma patients. FIG. 1A provides demographic, clinical, and molecular information for patients analyzed herein. Genomic alterations were obtained by tumor sequencing data generated for the PNOC003 trial, biorepository sequencing project, or other clinical studies. FIG. IB is a flow chart of biofluid samples analyzed herein. Patients were either enrolled in PNOC003 trial (DIPG) or Children's National (CN) brain tumor biorepository (MLG); *patients enrolled on 1339 to donate specimens at postmortem.
FIGS. 2A-N are graphical representations of genomic DNA validation for specific detection of mutant and wild type alleles of genes encoding driver mutations in pedatric diffuse midline gliomas. Shown are representative ddPCR plots for mutation detection using tumor genomic DNA from seven MLG patients and two non-CNS diseased brain tissue controls. FIGS. 2A-2G are from tumor tissue; FIGS. 2H-2N are from two non-CNS diseased brain tissue controls.
FIG. 3A-G are graphical representations of results obtained when assessing the sensitivity of a ddPCR platform with pre-amplification quantities of 2500 pg, 250 pg, 25 pg, and 2.5 pg of input DNA Genomic DNA. The DNA was obtained from frozen tumor tissue obtained at postmortem, from a H3.3 K27M mutant DIPG patient was diluted and used to assess for sensitivity. FIGS. 3A-3C are samples assessed without pre-amplification; FIGS. 3D-3G are from samples assessed with preamplification.
FIGS. 4A-4B are graphical representations assessing the specificity of s ddPCR platform by testing for H3.3 K27M in H3 wildtype DIPG tumor tissue and liquid bio me. FIG. 4A shows matched tumor tissue and CSF collected at postmortem from the same H3 WT DIPG patient who was negative for H3F3A p.K27M. FIG. 4B shows that plasma collected at initial diagnosis was negative for H3F3A p.K27M in three H3 wild type DIPG patients.
FIG. 5A-B show schematics of biofluid specimen processing for ddPCR analysis. FIG. 5A shows that genomic DNA was isolated from tumor tissue and used for ddPCR, while plasma and CSF were frozen upon collection, processed for ctDNA isolation, preamplified, and analyzed by ddPCR. FIG. 5B shows that circulating DNA isolated from biofluid specimens were analyzed in a three-step method by ddPCR, which includes dropletization (by a'Source' machine), PCR, followed by detection in the 'Sense' machine. An example is shown for H3F3A p.K27M detection, where LNA probes detect a single nucleotide change differentiating mutant and wild type alleles during PCR.
FIGS. 6A-F are graphic and pictorial representations showing detection of an histone 3 p.K27M mutation (H3K27M) ctDNA in biofluids of patients diagnosed with midline glioma. FIG. 6A shows detection of H3K27M ctDNA in CSF collected throughout a disease. FIG. 6B shows significantly higher ctDNA MAFs for H3K27M were detected in CSF compared to plasma. FIG. 6C shows higher H3K27M MAFs were present in CSF compared to plasma in the same patient. FIG. 6D shows that plasma ctDNA collected at diagnosis in DIPG patients enrolled in the PNOC003 clinical trial analyzed for H3K27M. Red asterisk denotes histone 3 wild type DIPGs. FIG. 6E shows that detection of H3K27M plasma ctDNA at post radiation in two patients that lacked detection at diagnosis. FIG. 6F shows an Increase in tumor volume by MRI at post radiation compared to diagnosis in patients shown in FIG. 6C.
FIGS. 7A-F show multiplexed detection of oncohistone and partner mutations in tumor and CSF of MLG patients. Genomic DNA and CSF ctDNA were analyzed from the same MLG patient for multiplexed detection of mutations.
FIGS. 8A-C show CSF collected from various neuroanatomical locations in MLGs. Bar graphs represent average for H3K27M MAF detection from technical duplicates, and error bars represent standard error of mean. FIG. 8A shows ignificantly higher H3K27M MAF was detected in CSF collected from adjacent sites to tumor compared to distant in MLG patients. FIG. 8B shows Cohort based analysis of CSF collected from DIPG patients from different neuroanatomical locations, indicating higher detection in locations closer to brainstem. FIG. 8C shows matched analysis for CSF collected from lateral ventricles and lumbar spine at postmortem from the same DIPG patient, showing similar trends as in (FIG. 8A) and (FIG. 8B).
FIGS. 9A-D show detection of a novel histone 3 mutation in fluid present in a brainstem tumor cyst found in a DIPG patient at postmortem. FIG. 9A is an image of a cyst (indicated by an arrow) in fresh tissue at postmortem. FIG. 9B is an MRI image captured a month before postmortem indicating cyst in pontine tumor. FIGS. 9C-9D show that higher MAF levels were found in cyst fluid (41%) compared to tumor tissue (34%), and CSF (38%) for H3F3A p.K27M. FIGS 10A-B shows detection of H3.3 K27M ctDNA in CSF at postmortem in a DIPG patient that tested negative in CSF collected at diagnosis. FIG. 10A shows detection of
H3K27M ctDNA in CSF collected at postmortem from a patient known to harbor H3.3 K27M in tumor. FIG.10B shows the results of an MRI imaging at diagnosis and postmortem for the same patient CSF was analyzed for in FIG. 10A.
FIGS. 11A-F show that a temporal analysis of plasma ctDNA matches response to therapy in DIPG patients. FIG. 11 A shows serial plasma ctDNA analysis of changes in H3K27M throughout course of treatment in patients that followed PNOC003 recommended therapy. FIG. 1 IB shows dynamic changes in plasma ctDNA and MRI tumor measurements in one patient in response to therapy. FLAIR and Tl-weighted post- gadolinium MRI images prior to treatment demonstrating a prominent expansile pontine mass and an additional focus within right cerebellar hemisphere that demonstrates evidence of enhancement. After treatment, the pontine and right cerebellar hemisphere lesions decreased in size. FIG. 11C shows fluctuations in plasma ctDNA and MRI tumor volumes during therapy. FIG. 1 ID shows a significant decrease in plasma ctDNA and MRI tumor volumes in response to radiation therapy. FIG. 1 IE shows a decrease in plasma ctDNA and MRI tumor burden from diagnosis to pre-cycle 3 of PNOC003 recommended therapy.
FIGS. 12A-12N show longitudinal changes in plasma ctDNA in association with MR imaging findings and clinical assessments. Each line graph represents plasma ctDNA and MRI changes in an individual patient diagnosed with DIPG. The red line depicts MRI tumor measurements and black line depicts changes in temporal plasma ctDNA. Error bars are standard error of mean for technical triplicates of plasma ctDNA assessed for MAF of H3K27M. The colored legend at the bottom of each figure indicates the time point of plasma ctDNA and MRI assessment during course of disease: green for initial diagnosis/biopsy, grey for during therapy, red for tumor growth, and black for the end of therapy following tumor growth/progression.
FIG. 13A-C are histograms showing bio fluid ctDNA and tumor spread as assessed by MRI, genomic, and/or histological studies. FIG. 13A shows tumor extension beyond site of primary tumor (pons or thalamus) as determined based on MRI obtained prior to postmortem, or molecular and/or histopathology of autopsied whole brain specimens. CSF ctDNA was higher in 18 MLG patients with tumor extension as compared to three MLG patients without tumor spread. Tumor involvement beyond pons was assessed by MRI review of patients enrolled in PNOC003. Plasma ctDNA and MRI collected from DIPG patients were analyzed (FIG. 13B) at initial diagnosis and (FIG. 13C) at time points during the course of disease (initial diagnosis, during therapy, and tumor growth). Unlike in CSF, ctDNA levels in plasma were not higher in DIPG patients with tumor spread.
DETAILED DESCRIPTION OF THE DISCLOSURE
The use of circulating tumor DNA (ctDNA) to monitor disease progression is non/minimally- invasive method that has been increasingly employed for disease monitoring in adult cancers including glioblastoma (GBM), melanoma, lung, breast, and colon cancersi3-n74 ,however such studies have not been applied to the pediatric population. Thus, there is an urgent need for the development of ctDNA assays for clinical applications in pediatric CNS patients.
Described herein is a sensitive, specific, rapid and minimally-invasive method for tumor surveillance of pediatric MLGs by monitoring the liquid biome. In embodiments, we show the detection of mutations associated with pediatric MLGs such as DIPG and thalamic tumors using patients' CSF and plasma. Further, using plasma collected through an ongoing clinical trial (PNOC003; NCT 2274987) conducted by the Pacific Pediatric Neuro-Oncology Consortium (PNOC), we show that mutation allelic frequency (MAF) is readily obtained from ctDNA present in liquid biome, and that MAF correlates with treatment response.
The disclosure provides a robust application of the use of ctDNA for tumor profiling, and assessment of tumor response to therapy in pediatric patients with MLGs. These results show the feasibility of incorporating liquid biopsy as a sensitive and minimally invasive tool to inform clinical management for children with MLGs
The use of circulating tumor DNA (ctDNA) to monitor disease progression is a noninvasive method that has been shown to be successful in adult glioblastoma, lung cancer, melanoma, breast cancer, and colon cancer (Tie et al. (2016) Sci. Transl. Med., 8(346):346ra92; Tsao et al. (2015) Sci. Rep., 5:11198, Garcia-Murillas et al. (2015) Sci Transl Med.,
7(302):302ral33; Schiovan et al. (2015) Sci. Transl. Med., 7(313):313ral82; De Mattos-Arruda et al. (2015) Nat. Commun., 6:8839). Several studies have reported the clinical utility of ctDNA as a biomarker for monitoring changes in tumor burden following therapy, resistance to therapy, and emergence of recurrence in melanoma, breast, lung, colon, ovarian cancer (Murtaza et al. (2013) Nature 497(7447): 108- 12; Tsao et al. (2015) Sci. Rep., 5:11198; Tie et al. (2016) Sci. Transl. Med., 8(346) :346ra92; Garcia-Murillas et al. (2015) Sci Transl Med., 7(302):302ral33). Longitudinal changes in ctDNA are a useful biomarker to monitor tumor progression in adult glioblastoma, and in patients with metastasis to the brain from breast and lung cancer (De Mattos-Arruda et al. (2015) Nat. Commun., 6:8839).
However, the validity of ctDNA as a biomarker in the clinical setting for assessing tumor response, longitudinal assessment of tumor size, tumor metastasis, and prediction of relapse is unknown. Moreover, given that ctDNA is detectable in less than 50% of primary brain tumors (Bettegowda et al. (2014) Sci Transl. Med., 6(224):224ra24) and that allelic frequency of H3F3A is low (Lewis et al. (2013) Science 340(6134): 857-61 ; Bender et al. (2013) Cancer Cell
24(5):660-72), a sensitive approach is needed for detection of such driver mutations in pediatric MLGs.
As described below, ctDNA was molecularly characterized in cerebrospinal fluid (CSF) and plasma, and the clinical utility of ctDNA to monitor tumor burden in pediatric midline gliomas (MLGs) was assessed. The disclosure shows that liquid-biopsy platforms can allow detection of ctDNA to reflect tumor regression or progression following therapeutic intervention.
Digital Droplet PCR
In embodiments, methods are performed using highly sensitive PCR-based sequencing method, such as digital droplet PCR.
As used herein, "digital PGR" refers to PCR wherein the sample is divided into discrete subunits prior to amplification by PGR. For example, the sample may be separated into thousands or millions of partitions, each containing either zero or one (or, at most, a few) template molecules. Fluorescent probes may be used to identify amplified target DNA in the partitions. Samples containing amplified product (e.g., fluorescent) are positive and those without product (e.g., no fluorescence) are negative. The ratio of positives to negatives in each sample can be used for quantification. For example, Poisson statistics can be used to determine the absolute template quantity without the need to consider the number of amplification cycles.
As used herein, "droplet digital PCR" (ddPCR) refers to digital PCR wherein the PCR solution is divided into smaller reactions (e.g., picoliter sized) through a water oil emulsion technique, which are then made to ran PCR individually. Typically, the PCR sample is partitioned into nanoliter-size samples and encapsulated in oil droplets, (see, e.g., Hinson et al. (2011) Anal. Chem., 83:8604-8610; Pinheiro et al. (2012) Anal. Chem.,
84:1003-1011).
The PCR process is well known in the art and includes, for example, methods described in US Patent No. 9,984,201, the contents of which are incorporated by reference herein in their entirety. These methods include, e.g., reverse transcription PCR, ligation mediated PCR, digital PCR (dPCR), or droplet digital PCR (ddPCR). For a review of PCR methods and protocols, see, e.g., Innis et al, eds., PCR Protocols, A Guide to Methods and Application, Academic Press, Inc., San Diego, Calif. 1990; U.S. Pat. No. 4,683,202 (Mullis). PCR reagents and protocols are also available from commercial vendors, such as Roche Molecular Systems.
In some embodiments, PCR is carried out as an automated process with a thermostable enzyme. In this process, the temperature of the reaction mixture is cycled through a denaturing region, a primer annealing region, and an extension reaction region automatically. Machines specifically adapted for this purpose are commercially available.
In some embodiments, amplified sequences are also measured using invasive cleavage reactions such as the Invader™ technology (Zou et al, 2010, Association of Clinical Chemistry (AACC) poster presentation on Jul. 28, 2010, "Sensitive Quantification of Methylated Markers with a Novel Methylation Specific Technology; and U.S. Pat. No. 7,011,944 (Prudent et al)).
Suitable next generation sequencing technologies are widely available. Examples include the 454 Life Sciences platform (Roche, Branford, Conn.) (Margulies et al. 2005 Nature, 437, 376-380); Ulumina's Genome Analyzer, GoldenGate Methylation Assay, or Infinium
Methylation Assays, i.e., Infinium HumanMethylation 27K BeadArray or VeraCode GoldenGate methylation array (Illumina, San Diego, Calif.; Bibkova et al, 2006, Genome Res. 16, 383-393; U.S. Pat. Nos. 6,306,597 and 7,598,035 (Macevicz); U.S. Pat. No. 7,232,656 (Balasubramanian et al.)); QX200.TM. Droplet DigitaLTM. PCR System from Bio-Rad; or DNA Sequencing by Ligation, SOLiD System (Applied Biosystems/Life Technologies; U.S. Pat. Nos. 6,797,470,
7,083,917, 7,166,434, 7,320,865, 7,332,285, 7,364,858, and 7,429,453 (Barany et al); the HeUcos True Single Molecule DNA sequencing technology (Harris et al, 2008 Science, 320, 106-109; U.S. Pat. Nos. 7,037,687 and 7,645,596 (Williams et al); U.S. Pat. No. 7,169,560 (Lapidus et al); U.S. Pat. No. 7,769,400 (Harris)), the single molecule, real-time (SMRT.TM.) technology of Pacific Biosciences, and sequencing (Soni and Meller, 2007, Clin. Chem. 53, 1996-2001);
semiconductor sequencing (Ion Torrent; Personal Genome Machine); DNA nanoball sequencing; sequencing using technology from Dover Systems (Polonator), and technologies that do not require amplification or otherwise transform native DNA prior to sequencing (e.g., Pacific Biosciences and Helicos), such as nanopore-based strategies (e.g., Oxford Nanopore, Genia Technologies, and Nabsys). These systems allow the sequencing of many nucleic acid molecules isolated from a specimen at high orders of multiplexing in a parallel fashion. Each of these platforms allow sequencing of clonally expanded or non-amplified single molecules of nucleic acid fragments. Certain platforms involve, for example, (i) sequencing by ligation of dye- modified probes (including cyclic ligation and cleavage), (ii) pyrosequencing, and (iii) single- molecule sequencing.
In an embodiment, methods are performed using digital droplet PCR (ddPCR). Among various methods of analyzing ctDNA derived from biofluids, digital droplet PCR (ddPCR) is one of the most sensitive methods compared to Sanger sequencing, quantitative PCR, and next generation sequencing techniques (Diaz et al. (2014) J. Clin. Oncol. 32(6):579-86; Baker, M. (2012) Nat Meth., 9:541-544).
After PCR amplification, the nucleic acids may be quantified by counting the sub- samples that contain PCR end-product (positive reactions) and the sub- samples containing no PCR end-product (negative reactions) taking into account the Poisson distribution. Digital PCR (dPCR) is, contrary to conventional PCR, not dependent on the number of amplification cycles performed in order to allow for a determination of the initial sample amount, thus eliminating the reliance on uncertain exponential data to quantify target nucleic acids and providing absolute quantification. In embodiments, "Digital droplet PCR" as used herein relates to a digital PCR method in which the initial sample is sub-divided into several droplets constituting the sub- samples.
Herein, ddPCR was used to detect major mutations in ctDNA of liquid biome specimens and to assess dynamic changes in ctDNA during a disease course. H3 27M partners with at least one additional driver mutation in cell cycle regulatory or growth factor signaling pathways (Nikbakht et al. (2016) Nat. Commun., 7:11185). Here, the presence of H3K27M and partner mutations in ctDNA was identified from liquid biome of MLGs. Significantly, the amount of detectable ctDNA in CSF and plasma was assessed with regard to clinical variables such as response to therapy, MRI tumor measurements, stage of disease, tumor spread, overall survival, and neuroanatomieal location of biofluid collection. More specifically, twenty- seven CSF samples were collected at various stages of disease (upfront, recurrence, and postmortem) from pediatric high grade midline gliomas. Upfront and longitudinal plasma samples (n=68) were collected with each MRI, from 22 diffuse intrinsic pontine glioma (DIPG) patients enrolled in a Phase I clinical trial. For monitoring etDNA, a liquid biopsy assay using a sensitive and robust digital droplet PGR system on CSF and plasma samples was developed. Major DIPG driver mutations including H3F3A (e.g., p. 27M),
HIST1H3B (e.g., p. 27M), ACVR1 (e.g., p.G328V/p.R206H), PPM ID (e.g., p.E525X),
PI 3R1 (e.g., p.K567E), and PI 3CA (e.g., p.H1047R) were detected. Samples can also be analyzed for IDH1 (e.g., p,R132H) and BRAF (e.g., p.V600E) mutations. Specifically, H3K27M was detected in 80% of CSF and upfront plasma samples in patients that harbor the histone 3 mutation in tumor. Higher levels of ctDNA were detected in CSF compared to plasma. Longitudinal analysis of plasma showed a significant decrease (p = 0.005) in ctDNA following radiotherapy, whereas MRI tumor measurements did not show a significant decrease in tumor size (n=15). Furthermore, H3K27M was detected in ctDNA of plasma samples in xenograft models of DIPG. Thus, biofluids from patients and animal models (e.g., urine, blood, serum, plasma, CSF) is a suitable medium for detection and quantification of ctDNA. A liquid biopsy can complement or in some cases eliminate the need for biopsies and inform recurrence and tumor response to treatment.
In accordance with the instant invention, methods of treating cancer in a subject in need thereof are pro vided. In a particular embodiment, the method comprises determining the presence of a mutation in histone H3 (e.g., histone H3.3) and administering a therapy (e.g., administering at least one therapeutic agent to the subject) to the subject if the mutation in histone H3 is present. The method may further comprise the step of obtaining a biological sample from the subject. The biological sample of the methods may be a fluid or liquid such as blood, CSF, plasma, or urine. In a particular embodiment, the biological sample is blood or plasma. In a particular embodiment, the therapy administered to the subject is an epigenetic regulating drug such as, without limitation, SAHA and panobiiiostat.
The cancer detected and treated by the instant methods may be a brain tumor. More specifically, the cancer may be a pediatric brain tumor. In a particular embodiment, the cancer is a glioma, particularly a diffuse glioma or a diffuse intrinsic pontine glioma (DIPG). The methods of the instant invention may comprise detenrdning the presence of a mutation(s) in circulating tumor DNA (ctDNA). In a particular embodiment, determining of the presence of the mutation(s) comprises perforating digital PCR. In a particular
embodiment, the determining of the presence of the mutation comprises performing droplet digital PCR (ddPCR). The digital PCR or ddPCR may be performed with at least one primer comprising any one of SEQ ID NOs: 1-18. The digital PCR or ddPCR may be performed with primers comprising each of SEQ ID NOs: 1-18. The digital PCR or ddPCR may be performed with at least one probe comprising any one of SEQ ID NOs: 19-36. The digital PCR or ddPCR may be performed with probes comprising each of SEQ ID NOs: 19-36.
As stated herein above, the instant methods comprise determining the presence of a mutation in histone H3 (e.g., histone H3.3). In a particular embodiment, the mutation in histone H3 is K27M. In a particular embodiment, the histone H3 is H3F3A. In a particular
embodiment, the method comprises determining the presence of a mutation in H3F3A and at least one other gene (e.g., partner gene). The other gene(s) may be, for example, selected from the group of HIST1H3B (e.g., p.K27M), HIST2H3C (e.g., p.K27M), ACVR1 (e.g., p.G328V/p.R206H), PPM1D (e.g., p.E525X), PIK3R1 (e.g., p.K567E), PIK3CA (e.g., p.H1047R), IDH1 (e.g., p.R132H; isocitrate dehydrogenase (NADP(+)) 1, cytosolic; Gene ID: 3417) and BRAF (e.g., p.V600E; B-Raf proto-oncogene, serine/threonine kinase; Gene ID: 673) (e.g., 1, 2, 3, 4, 5, 6, 7, or all 8 genes are analyzed). In a particular embodiment, the other gene(s) is selected from the group of HIST1H3B (e.g., p.K27M), ACVR1 (e.g.,
p.G328V/p.R206H), PPM1D (e.g. p.E525X), PIK3R1 (e.g., p.K567E), and PIK3CA (e.g., p.H1047R) (e.g., 1, 2, 3, 4, or all 5 genes are analyzed). In a particular embodiment, the other gene(s) is selected from the group of HIST1H3B (e.g., p.K27M), ACVR1 (e.g.,
p.G328V/p.R206H), PPM1D (e.g., p.E525X), and PIK3R1 (e.g., p.K567E) (e.g., 1, 2, 3, or all 4 genes are analyzed). Sequence information for certain of these genes is provided below.
H3 histone family member 3A (H3F3A; SEQ ID NO: 37) Gene ID: 3020, Gen Bank Accession Nos. NM_002107.4 and NP_002098.1 (Note: the Met at position 1 is not included in amino acid numbering)
1 ARTKQTARK STGGKAPRKQ LATKAARKSA PSTGGVKKPH RYRPGTVALR EIRRYQKSTE 61 LLIRKLPFQR LVREIAQDFK TDLRFQSAAI GALQEASEAY LVGLFEDTNL CAIHAKRVTI
121 PKDIQLARR IRGERA Histone cluster 1 H3 family member b (HIST1H3B; SEQ ID NO: 38) Gene ID: 8358, Gen Bank Accession Nos. NM_003537.3 and NP_003528.1 (Note: the Met at position 1 is not included in amino acid numbering).
1 ARTKQTARK STGGKAPRKQ LATKAARKSA PATGGVKKPH RYRPGTVALR EIRRYQKSTE
61 LLIRKLPFQR LVREIAQDFK TDLRFQSSAV ALQEACEAY LVGLFEDTNL CAIHAKRVTI
121 PKDIQLARR IRGERA
Activin A receptor type 1 (ACVRl ; SEQ ID NO: 39) Gene ID: 90, Gen Bank Accession
NMJXJl 105.4 and NP_001096.1
1 MVDGVMILPV LIMIALPSPS MEDEKPKVNP KLYMCVCEGL SCGNEDHCEG QQCFSSLSIN
61 DGFHVYQKGC FQVYEQGKMT CKTPPSPGQA VECCQGDWCN RNITAQLPTK GKSFPGTQNF
121 HLEVGLIILS WFAVCLLAC LLGVALRKFK RRNQERLNPR DVEYGTIEGL ITT VGDSTL
181 ADLLDHSCTS GSGSGLPFLV QRTVARQITL LECVGKGRYG EVWRGSWQGE NVAV IFSSR
241 DEKSWFRETE LYNTVMLRHE NILGFIASDM TSRHSSTQLW LITHYHEMGS LYDYLQLTTL
301 DTVSCLRIVL SIASGLAHLH IEIFGTQGKP AIAHRDLKSK ILVKKNGQC CIADLGLAVM
361 HSQSTNQLDV GNNPRVGTKR YMAPEVLDET IQVDCFDSYK RVDIWAFGLV LWEVARRMVS
421 NGIVEDYKPP FYDWPNDPS FEDMRKWCV DQQRPNIPNR WFSDPTLTSL AKLM ECWYQ
481 NPSARLTALR IKKTLTKIDN SLDKLKTDC
Protein phosphatase, Mg2+/Mn2+ dependent ID (PPM1D; SEQ ID NO: 40) Gene ID: 84, Gen Bank Accession Nos. NM„003620.3 and NPJ303611.1
1 MAGLYSLGVS VFSDQGGRKY MEDVTQIWE PEPTAEEKPS PRRSLSQPLP PRPSPAALPG
61 GEVSGKGPAV AAREARDPLP DAGASPAPSR CCRRRSSVAF FAVCDGHGGR EAAQFAREHL
121 WGFIKKQKGF TSSEPAKVCA AIRKGFLACH LAMWKKLAEW PKTMTGLPST SGTTASWII
181 RGMKMYVAHV GDSGWLGIQ DDPKDDFVRA VEVTQDHKPE LPKERERIEG LGGSVMNKSG
241 VNRWWKRPR LTHNGPVRRS TVIDQIPFLA VARALGDLWS YDFFSGEFW SPEPDTSVHT
301 LDPQKHKYII LGSDGLWNMI PPQDAISMCQ DQEEKKYLMG EHGQSCAKML V RALGRWRQ
361 RMLRADNTSA IVICISPEVD NQGNFTNEDE LYLNLTDSPS Y SQETCVMT PSPCSTPPV
421 SLEEDPWPRV NSKDHIPALV RS AFSENFL EVSAEIAREN VQGWIPS D PEPLEENCAK
481 ALTLRIHDSL SLPIGLVP TNSTNTV DQ K LKMSTPGQ MKAQEIERTP PTNFKRTLEE
541 SNSGPLMKKH RRNGLSRSSG AQPASLPTTS QRKNSVKLTM RRRLRGQKKI GNPLLHQHRK
601 TVCVC
Phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1 ; SEQ ID NO: 41) Gene ID: 529 Gen Bank Accession Nos. NM_181523.2 and NP_852664.1
1 MSAEGYQYRA LYDYKKEREE DIDLHLGDIL TV KGSLVAL GFSDGQEARP EEIGWLNGYN
61 ETTGERGDFP GTYVEYIGRK KISPPTPKPR PPRPLPVAPG SSKTEADVEQ QALTLPDLAE
121 QFAPPDIAPP LLIKLVEAIE KKGLECSTLY RTQSSSNLAE LRQLLDCDTP SVDLEMIDVH
181 VLADAFKRYL LDLPNPVIPA AVYSEMISLA PEVQSSEEYI QLLKKLIRSP SIPHQYWLTL
241 QYLLKHFFKL SQTSSKNLL ARVLSEIFSP MLFRFSAASS DNTENLIKVI EILISTEWNE
301 RQPAPALPPK PPKPTTVAN GMN NMSLQD AEWYWGDISR EEVNEKLRDT ADGTFLVRDA
361 STKMHGDYTL TLRKGGNNKL IKIFHRDGKY GFSDPLTFSS WELINHYR ESLAQY PKL
421 DVKLLYPVSK YQQDQWKED NIEAVGKKLH EYNTQFQE S REYDRLYEEY TRTSQEIQMK
481 RTAIEAFNET IKIFEEQCQT QERYSKEYIE KFKREGNEKE IQRIMHNYDK LKSRISEIID
541 SRRRLEEDLK KQAAEYREID KRMNSI PDL IQLRKTRDQY LMWLTQKGVR QKKLNE LGN 601 ENTEDQYSLV EDDEDLPHHD EKTWNVGSSN K KAENLLRG KRDGTFLVRE SSKQGCYACS
661 WVDGEVKHC VINKTATGYG FAEPYNLYSS LKELVLHYQH TSLVQHNDSL VTLAYPVYA
72 QQRR
Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA; SEQ ID NO: 42) Gene ID: 5290, Gen Bank Accession Nos. NM_006218.3 and NP_006209.2
1 MPPRPSSGEL WGIHLMPPRI LVECLLPNGM IVTLECLREA TLITIKHELF KEARKYPLHQ
61 LLQDESSYIF VSVTQEAERE EFFDETRRLC DLRLFQPFLK VIEPVGNREE KILNREIGFA
121 IG PVCEFDM VKDPEVQDFR RNILNVCKEA VDLRDLNSPH SRAMYVYPPN VESSPELPKH
181 IYNKLDKGQI IWIWVIVSP NNDKQKYTLK INHDCVPEQV IAEAIRKKTR SMLLSSEQLK
241 LCVLEYQGKY ILKVCGCDEY FLEKYPLSQY KYIRSCIMLG RMPNLML AK ESLYSQLPMD
301 CFTMPSYSRR ISTATPY NG ETSTKSLWVI NSALRIKILC ATYVNVNIRD IDKIYVRTGI
361 YHGGEPLCDN VNTQRVPCSN PRWNEWLNYD IYIPDLPRAA RLCLSICSVK GRKGAKEEHC
421 PLAWGNINLF DYTDTLVSGK ALNLWPVPH GLEDLLNPIG VTGSNPNKET PCLELEFDWF
481 SSWKFPDMS VIEEHANWSV SREAGFSYSH AGLSNRLARD NELRENDKEQ LKAISTRDPL
541 SEITEQEKDF LWSHRHYCVT IPEILPKLLL SVKWNSRDEV AQMYCLVKDW PPIKPEQAME
601 LLDCNYPDPM VRGFAVRCLE KYLTDDKLSQ YLIQLVQVLK YEQYLDNLLV RFLLKKALTN
661 QRIGHFFFWH LKSEMHNKTV SQRFGLLLES YCRACGMYLK HLNRQVEAME KLINLTDILK
721 QEKKDETQKV Q KFLVEQ R RPDFMDALQG FLSPLNPAHQ LGNLRLEECR IMSSAKRPLW
781 LNWENPDIMS ELLFQNNEII FKNGDDLRQD MLTLQIIRIM ENIWQNQGLD LRMLPYGCLS
841 IGDCVGLIEV VRNSHTIMQI QCKGGLKGAL QFNSHTLHQW LKDKNKGEIY DAAIDLFTRS
901 CAGYCVATFI LGIGDRHNSN IMVKDDGQLF HIDFGHFLDH KKKKFGYKRE RVPFVLTQDF
961 LIVISKGAQE CTKTREFERF QEMCYKAYLA IRQHANLFIN LFS MLGSGM PELQSFDDIA
1021 YIRKTLALDK TEQEALEYFM KQ NDAHHGG WTTKMDWIFH TIKQHALN
In accordance with another aspect of the instant invention, methods of determining the efficacy of a cancer therapy are provided. In a particular embodiment, the method comprises 1) administering a cancer therapy to a subject in need thereof, 2) obtaining a biological sample from the subject after administration of the cancer therapy to the subject, and 3) determining the presence of a mutation in in the biological sample (e.g., in histone H3 (e.g., histone H3.3)). The method may comprise obtaining multiple biological samples over time from the subject after administration of the cancer therapy to the subject. The method may comprise obtaining a biological sample from the subject before administration of the cancer therapy to the subject (e.g., a baseline). A decrease in the presence of the mutation (e.g., in histone H3) over time (e.g., when multiple biological samples are obtained after therapy) or compared to the amount observed prior to therapy, indicates that the cancer therapy administered to the subject is effective (e.g., inhibiting tumor growth).
The biological sample of the methods may be a fluid or liquid such as blood, CSF, plasma, serum, or urine. In a particular embodiment, the biological sample is blood or plasma. The cancer monitored by the instant methods may be a brain tumor. More specifically, the cancer may be a pediatric brain tumor. In a particular embodiment the cancer is a glioma, particularly a diffuse glioma or a diffuse intrinsic pontine glioma (DIPG).
The cancer therapy administered to the subject may comprise any type of therapy. For example, the cancer therapy may comprise the administration of at least one chemotherapeutie agent (e.g., a small molecule). The cancer therapy may include radiation therapy.
The methods of the instant invention may comprise determining the presence of the mutation in circulating tumor DNA (ctDNA). In a particular embodiment, determining of the presence of the mutation comprises performing digital PGR. In a particular embodiment, the determining of the presence of the mutation comprises performing droplet digital PGR (ddPCR). The digital PGR or ddPCR may be performed with at least one primer comprising any one of SEQ ID NOs: 1-14 or a portion thereof. The digital PGR or ddPCR may be performed with at least one probe comprising any one of SEQ ID NOs: 19-36. The digital PGR or ddPCR may be performed with probes comprising each of SEQ ID NOs: 19-36.
As stated hereinabove, the instant methods comprise determining the presence of a mutation (e.g., in histone H3, such as histone H3.3). In a particular embodiment, the method comprises determining the presence of a mutation in a gene(s) selected from the group of H3F3 A (e.g., p.K27M), HIST1H3B (e.g., p.K27M), HIST2H3C (e.g., p.K27M), ACVR1 (e.g., p.G328V/p.R206H), PPM1 (e.g., p.E525X), FK3R1 (e.g., p. 567E), PIK3CA (e.g., p.H1047R), IDH1 (e.g., p.R132H; isocitrate dehydrogenase (NADP(+)) 1, cytosolic (Gene ID:3417)) an
BRAF (e.g., p.V600E; B-Raf proto-oncogene, serine/threonine kinase (Gene ID: 673)) (e.g., 1, 2, 3, 4, 5, 6, 7, or all 8 genes are analyzed). In a particular embodiment, the gene(s) is selected from the group of H3F3A (e.g., p.K27M), HIST1H3B (e.g., p.K27M), ACVR1 (e.g.,
p.G328V/p.R2()6H), PPM ID (e.g., p.E525X), PIK3R1 (e.g., p. 567E), and PI 3CA (e.g., p.H1047R) (e.g., 1, 2, 3, or all 5 genes are analyzed). In a particular embodiment, the other gene(s) is selected from the group of HIST1H3B (e.g., p.K27M), ACVR1 (e.g.,
p.G328V/p.R206H), PPM1D (e.g., p.E525X), and PIK3R1 (e.g., p.K567E) (e.g.,2, 3, or all 4 genes are analyzed). In a particular embodiment, the mutation in histone H3 is K27M. In a particular embodiment, the histone H3 is H3F3A. In a particular embodiment, the method comprises determining the presence of a mutation in H3F3A and at least one other gene. A decrease in the presence of the mutations over time (e.g., when multiple biological samples are obtained after therapy) or compared to the amount observed prior to therapy, indicates that the cancer therapy administered to the subject is effective (e.g., inhibiting tumor growth).
OUgonucleotide probes are also provided with the instant invention. The probes are designed to have high affinity and specificity to the target site (e.g., the mutations set forth herein and, optionally, the wild- type gene). In a particular embodiment, oligonucleotide probe(s) target a gene (e.g., wild-type and/or mutant) selected from the group of H3F3A (e.g., p.K27M), HIST1H3B (e.g., p.K27M), HIST2H3C (e.g., p.K27M), ACVR1 (e.g., p.G328V/p.R206H), PPM1D (e.g., p.E525X), PIK3R1 (e.g., p.K567E), PIK3CA (e.g., p.H1047R), IDH1 (e.g., p.R132H) and BRAF (e.g., p.V600E).
The probes do not have an absolute requirement on length. However, the probes will typically be from about 10 to about 250 nucleotides, about 10 to about 100, about 10 about 50 nucleotides, about 10 to about 40 nucleotides, about 10 to about 30 nucleotides, about 10 to about 25 nucleotides, or about 10 to about 20 nucleotides. In a particular embodiment, the probe is at least about 10 nucleotides in length. The probe may be at least 85%, at least 90%, at least 95%, at least 97%, or, more preferably, 100% complementary to the target sequence. In a particular embodiment, the oligonucleotide probe is designed such that the mutation is towards the middle of the sequence of the probe (e.g., within the middle third of the probe length).
In a particular embodiment, the probe may comprise at least one nucleotide analog. For example, the nucleotide analogs may be used to increase annealing affinity and/or specificity and/or resistance to degradation. For example, the use of locked nucleic acid (LNA) bases within an oligonucleotide increases the affinity and specificity of annealing of the oligonucleotide to its target site (Kauppinen et al.(2005) Drug Discov. Today Tech., 2:287-290; Orum et al. (2004) Letters Peptide Sci., 10:325-334). Nucleotide analogs include, without limitation, nucleotides with phosphate modifications comprising one or more phosphorothioate, phosphorodithioate, phosphodiester, methyl phosphonate, phosphoramidate, methylphosphonate, phosphotriester, phosphoroaridate, morpholino, amidate carbamate, carboxymethyl, acetamidate, polyamide, sulfonate, sulfonamide, sulfamate, formacetal, thioformacetal, and/or alkylsilyl substitutions; nucleotides with modified sugars; and nucleotide mimetics such as, without limitation, peptide nucleic acids (PNA), morpholino nucleic acids, cyclohexenyl nucleic acids, anhydrohexitol nucleic acids, glycol nucleic acid, threose nucleic acid, and locked nucleic acids (LNA). In a particular embodiment, the probes comprise at least one locked nucleic acid. The probes may comprise one of SEQ ID NOs: 19-36 or a sequence with at least 85%, at least 90%, at least 95%, or at least 97% identity to one of SEQ ID NOs: 19-36. In a particular embodiment, the probe comprises one of SEQ ID NOs: 19-36. The probes may comprise additional nucleotides 5' or 3' to the included SEQ ID NO. For example, the probe may comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9,10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more nucleotides 5' or 3' to the included SEQ ID NO. In a particular embodiment, the additional sequences are complementary to the target sequence.
The probes of the instant invention may comprise one or more fluorescent probes / fluorophores and/or quenchers. The fluorophores and or quenchers may be added to the 5' and or 3' termini of the probes. The fluorophores and/or quenchers may also be added to internal part of the probes (e.g., a ZEN probe). Fluorophores and/or quenchers are well known in the art (see, e.g., IDT, Coralville, IA). Examples of fluorophores and/or quenchers include, without limitation, 6-FAM, fluorescein, Cy3, Cy5, TAMRA, JOE, MAX, TET, Cy5.5, ROX, ATTO, TYE, Yakima Yellow®, HEX, TEX, Texas Red®, Iowa Black®, ZEN, and Alexa Fluor®. The fluorophores and/or quenchers used allow for the determination of the presence of the wild- type and/or mutant allele in a sample at the same time (see, e.g., the Examples). In a particular embodiment, the fluorophores and/or quenchers create an energy transfer pair (e.g., fluorescence resonance energy transfer (FRET)) (e.g.,as set forth in Table 1). In a particular embodiment, the probes comprise a fluorophore and/or quencher combination presented in Table 1. In a particular embodiment, the probe comprises any one of SEQ ID NOs: 19-36. In a particular embodiment, the probe comprises any one of SEQ ID NOs: 19-36 along with the modifications presented in Table 1.
Compositions comprising at least one probe of the instant invention are also provided. In a particular embodiment, the composition is an aqueous solution. In a particular embodiment, the composition comprises at least one probe comprising any one of SEQ ID NOs: 19-36. In a particular embodiment, the composition comprises individual probes comprising each of SEQ ID NOs: 19-36.
Compositions comprising at least one primer of the instant invention are also provided. In a particular embodiment, the composition is provided an aqueous solution. In a particular embodiment, the composition comprises at least one primer comprising any one of SEQ ID NOs: 1-18. In a particular embodiment, the composition comprises individual primers comprising each of SEQ ID NOs: 1-18.
Definitions
The following definitions are provided to facilitate an understanding of the present invention:
The singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise.
"Pharmaceutically acceptable" indicates approval by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
A "carrier" refers to, for example, a diluent, adjuvant, preservative (e.g.,Thimersol, benzyl alcohol), anti-oxidant (e.g., ascorbic acid, sodium metabisulfite), solubilizer (e.g., polysorbate 80), emulsifier, buffer (e.g., Tris HC1, acetate, phosphate), antimicrobial, bulking substance (e.g., lactose, mannitol), excipient, auxiliary agent or vehicle (e.g., with which an active agent of the present invention is administered). Pharmaceutically acceptable carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin. Water or aqueous saline solutions and aqueous dextrose and glycerol solutions may be employed as carriers, particularly for injectable solutions. Suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin (Mack Publishing Co., Easton, PA); Gennaro, A. R., Remington: The Science and Practice of
Pharmacy, (Lippincott, Williams and Wilkins); Liberman, et al., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y.; and Kibbe, et al., Eds., Handbook of Pharmaceutical Excipients, American Pharmaceutical Association, Washington.
As used herein, the term "small molecule" refers to a substance or compound that has a relatively low molecular weight (e.g., less than 4,000, less than 2,000, particularly less than 1 kDa or 800 Da). Typically, small molecules are organic, but are not proteins, polypeptides, or nucleic acids, though they may be amino acids or dipeptides.
The term "treat" as used herein refers to any type of treatment that imparts a benefit to a patient afflicted with a disease, including improvement in the condition of the patient (e.g., in one or more symptoms), delay in the progression of the condition, etc. As used herein, the term "prevent" refers to the prophylactic treatment of a subject who is at risk of developing a condition resulting in a decrease in the probability that the subject will develop the condition.
As used herein, "diagnose" refers to detecting and identifying a disease or disorder in a subject. The term may also encompass assessing or evaluating the disease or disorder status (progression, regression, stabilization, response to treatment, etc.) in a patient known to have the disease or disorder.
As used herein, the term "prognosis" refers to providing information regarding the impact of the presence of a disease or disorder (e.g., as determined by the diagnostic methods of the present invention) on a subject's future health (e.g., expected morbidity or mortality, the likelihood of getting or risk of cholestasis). In other words, the term
"prognosis" refers to providing a prediction of the probable course and outcome of a disease/disorder and/or the likelihood of recovery from the disease/disorder.
As used herein, the term "subject" refers to an animal, particularly a mammal, particularly a human.
A "therapeutically effective amount" of a compound or a pharmaceutical
composition refers to an amount effective to prevent, inhibit, treat, or lessen the
symptoms of a particular disorder or disease. The treatment of a disease or disorder herein may refer to curing, relieving, and/or preventing the disease or disorder, the symptom(s) of it, or the predisposition towards it.
As used herein, the term "therapeutic agent" refers to a chemical compound or biological molecule including, without limitation, nucleic acids, peptides, proteins, and antibodies that can be u ed to treat a condition, disease, or disorder or reduce the symptoms of the condition, disease, or disorder.
The term "isolated" refers to the separation of a compound from other components present during its production or from its natural environment. "Isolated" is not meant to exclude artificial or synthetic mixtures with other compounds or materials, or the presence of impurities that do not substantially interfere with the fundamental activity, and that may be present, for example, due to incomplete purification, or the addition of stabilizers.
As used herein, a "biological sample" refers to a sample of biological material obtained from a subject, particularly a human subject, including a tissue, a tissue sample, cell(s), and a biological fluid (e.g., blood (e.g., whole blood), serum, plasma, urine, sweat, tears, saliva, mucosal secretions, sputum, CSF).
The term "probe" as used herein refers to an oligonucleotide, polynucleotide or nucleic acid, either RNA or DNA, which is capable of annealing with or specifically hybridizing to a nucleic acid with sequences complementary to the probe. A probe may be either single- stranded or double- stranded. The exact length of the probe will depend upon many factors, including temperature, source of probe and use of the method. For example, the oligonucleotide probe typically contains about 10-250, about 10-100, about 10-50, about 15-30, about 15-25, or about 10-20 nucleotides. The probes herein may be selected to be complementary to different strands of a particular target nucleic acid sequence. This means that the probes must be sufficiently complementary so as to be able to "specifically
hybridize" or anneal with their respective target strands under a set of pre-determined conditions. Therefore, the probe sequence need not reflect the exact complementary sequence of the target, although they may. For example, a non-complementary nucleotide fragment may be attached to the 5' or 3' end of the probe, with the remainder of the probe sequence being complementary to the target strand. Alternatively, non- complementary bases or longer sequences can be interspersed into the probe, provided that the probe sequence has sufficient complementarity with the sequence of the target nucleic acid to anneal therewith specifically. A probe may be tagged or labeled (i.e., attached to an entity making it possible to identify a compound to which it is associated (e.g., fluorescent or radioactive tag). In certain embodiments, a label is selected from the group consisting of biotin, copper-DOTA, biotin-PEG3, aminooxyacetate, 19FB, 18FB, FITC-PEG3, fluorescein and fluorescein derivatives (e.g., 5-carboxy fluorescein). In other embodiments, the label is selected from the group consisting of 64Cu DOT A, 68Ga DOT A, 18F, ^C , 68Ga, 89Zr, i24I, 86Y, 94mTc, n0raXn, "C and 76Br.
The term "primer" as used herein refers to an oligonucleotide, either RN A or DNA, either single- stranded or double-stranded, which, when placed in the proper environment, is able to functionally act as an initiator of template-dependent nucleic acid synthesis. When presented with an appropriate nucleic acid template, suitable nucleoside triphosphate precursors of nucleic acids, a polymerase enzyme, suitable cofactors and conditions such as appropriate temperature and H, the primer may be extended at its 3' terminus by the addition of nucleotides by the action of a polymerase or similar activity to yield a primer extension product. The primer may vary in length depending on the particular conditions and
requirement of the application. For example, the oligonucleotide primer is typically about 10- 25 or more nucleotides in length, but can be significantly longer. The primer must be of sufficient complementarity to the desired template to prime the synthesis of the desired extension product, that is, to be able to anneal with the desired template strand in a manner sufficient to provide the 3' hydroxyl moiety of the primer in appropriate juxtaposition for use in the initiation of synthesis by a polymerase or similar enzyme. It is not required that the primer sequence represent an exact complement of the desired template, though it may. For example, a non- complementary nucleotide sequence may be attached to the 5' end of an otherwise complementary primer. Alternatively, non-complementary bases may be interspersed within the oligonucleotide primer sequence, provided that the primer sequence has sufficient complementarity with the sequence of the desired template strand to functionally provide a template-primer complex for the synthesis of the extension product.
The term "oligonucleotide," as used herein refers to sequences, primers and probes of the present invention, and is defined as a nucleic acid molecule comprised of two or more ribo- or deoxyribonucleotides, preferably more than three. The exact size of the oligonucleotide will depend on various factors and on the particular application and use of the oligonucleotide.
The respective fluorophores are added to the 5' end of the probes, and the quenchers are added to the middle and at the 3' end of the probe. The symbol + following a nucleotide indicates a locked nucleic acid. IABkFQ: Iowa Black® FQ. SEQ ID NO is provided in parentheses. The SEQ ID NOs represent the nucleotide sequences without the probes or modifications.
Figure imgf000022_0001
Figure imgf000023_0001
Table 1: Primers & Probes for ddPCR
cDNA Mutation Forward Reverse Wild type Mutant
Gene
change (protein) Primer Primer probe probe
156-
5'- /5TET/TCGA
5'-CCAGACA FAM/TCGAG
ACCTCAGATA GAT+TTC+A
ACTGTTCAA AT+TTC+TC
BRAF 1799T>A V600E TATTTCTTCAT CT+GTAGCT
AC-3' T+GTAGCT/3
G-3' /3IABkFQ/
(18) IABkFQ/
(17) (35)
(36)
The following non- limiting examples are provided to illustrate various embodiments of the present invention. EXAMPLES
Example 1 : Methods
This example illustrates a sensitive and specific platform for discriminating rare, low abundant, tumor-associated circulating DNA in pediatric patients with mid-line glioma (MLG) tumors. Clinicopathological and genomic characteristics of the tumors of a pediatric MLG patient cohort are shown in Fig. 1A. Patients were diagnosed with MLGs harboring various mutation combinations, including oncohistone variants: 94% harbored histone 3 mutations (79% with H3.3K27M, 15% with H3.1K27M), and 6% were H3 wild type. The initial goal was to develop and validate a clinically relevant and minimally invasive Uquid biopsy platform suitable for detection and quantification of somatic mutations associated with pediatric MLGs. To address this, we validated our ddPCR probes for assessing mutation burden by using tumor genomic DNA from MLG patients harboring oncohistone H3F3A (p.K27M), HIST1H3B
(p.K27M) and partner mutants in genes ACVR1 (p.G328V, p.R206H), PPM1D (p.E525X), PIK3R1 (p.K567E), and BRAF (p.V600E) (FIGS. 2A-N). Additionally, no mutant clusters were observed where control brain genomic DNA was used as template (FIGS. 2A-N). To assess the sensitivity, we used a range (2.5ng, 250pg, 25pg and 2.5pg) of genomic DNA as template for ddPCR and detected a corresponding linear decrease (10 fold) in MAF, indicating a low (2.5pg) amount of DNA to be suitable for detection by ddPCR (FIGS. 3A-G).
To assess the specificity of our platform, we used a large cohort of control, non-CNS malignant specimens (CSF = 16; plasma = 20) and probed for wild type and mutant histone ctDNA. Our data determined that any allelic frequency detected as equal or below 0.001% to be false positive in the case of histone p.K27M analysis (Table 2). We also analyzed genomic DNA, CSF, and plasma obtained from DIPG patients who did not harbor histone 3 p.K27M mutations. No histone 117 mutant copies were detected in any of the specimen tested, indicating the specificity of our platform (FIGS. 4A-4B).
Example 2: CSF and plasma harbor circulating tumor DNA indicative of driver mutations associated with pediatric MLGs.
Liquid biome specimens were analyzed from 84 subjects (48 MLG patients, and 36 non- CNS diseased controls), enrolled in an ongoing clinical trial PNOC003 (NCT 227498), and consented for the Children's National (CN) brain tumor biorepository (FIG. IB). A cohort of 110 biofluid samples (30 CSF, 79 plasma, and 1 cyst fluid) representing 48 MLG patients was processed for liquid profiling (FIGS. 5A-B). CSF samples were collected at a single time point through the CN biorepository at pre-treatment, during therapy, and at postmortem from 27 MLG patients, while serial sampling at pre-treatment and postmortem was available for one patient with DIPG. Plasma specimens were obtained starting at diagnosis, and longitudinally throughout treatment from patients with DIPG enrolled in PNOC003 (NCT 2274987) (at initial biopsy n=23, during treatment n=15) (FIG. IB, Table 2). Three of the DIPG patients enrolled in PNOC003 also consented for CSF collection at postmortem via the CN biorepository. Two patients harbored H3 K27M and one was H3 wild type (FIG 1).
The results are shown in Table 2, which provides midline glioma patient demographic information, and samples analyzed for genomic and ctDNA studies. Genomic alterations were determined by sequencing conducted for PNOC003 clinical trial and biorepository sequencing study. Validation by ddPCR analysis of tumor samples is indicated in the table. EOT denotes end of therapy time point.
Histone 3 mutant and wild type alleles were detected in 75% of CSF specimens collected at diagnosis, 67% of those collected during treatment, and 90% of those collected at postmortem (Fig. 2a). H3 K27M- mutant ctDNA was detected in 89% of all CSF specimens analyzed from 27 MLGs, where 23 of these were confirmed to harbor oncohistones as assessed by tumor DNA analysis.
We have previously shown that in addition to oncohistone mutations, MLGs harbor obligate partner mutations in genes such as ACVR1, TP53, PPM ID, and PIK3R1. We thus sought to determine whether biofluids represent a source for detection of multiple mutations for a particular patient. We first used tumor DNA known to harbor oncohistone and partner mutations (ACVR1 p.R206H, PIK3R1 p.K567E, or BRAF p.V600E) to optimize the experimental and analytical approach for multiplexed assays (FIGS. 7A-F). We then tested CSF obtained from these patients and showed the feasibility of detecting mutant and wild type alleles, for oncohistone and obligate partners in ACVR1, PIK3R1 or BRAF (FIGS. 7A-F). Given the paucity of tumor DNA, and the invasive nature of surgical biopsy, our multiplexed mutation analysis allows for maximized clinical utility of a single accessible biofluid specimen.
We then assessed the effect of CSF location on ctDNA abundance. CSF collected from lateral ventricles, lumbar spine, cervical spine, subdural (at biopsy), ventriculoperitoneal (VP) shunt, and the fourth ventricle (cisternae magna tap) was analyzed by ddPCR. While we detected a significantly higher mutation allelic frequency in CSF collected from adjacent locations compared to distant, we report successful detection of H3 K27M ctDNA in all locations tested (FIGS. 8A-C). We previously reported the suitability of the cyst fluid for identification of tumor associated proteins. Here, we analyzed tumor cyst fluid, CSF, and tumor genomic DNA obtained at postmortem from a patient diagnosed with DIPG. We showed that the cyst fluid is highly enriched for tumor DNA as suggested by the high number of histone mutant DNA copies (FIGS. 9A-D).
To assess the enrichment of ctDNA in CSF versus plasma, we tested these two biofluids and found that in general, a significantly higher allelic frequency of ctDNA was detected in CSF (n=27 patients, 30 specimens) compared to plasma (n=20, 77 specimens) (p < 0.0001), which may indicate ctDNA enrichment in CSF due to proximity to the tumor mass (FIG. 6B). This cohort-based result was confirmed by our matched analysis (n=2), where we found an enrichment of ctDNA in CSF compared to plasma within the same patient (FIG. 6C). Our data indicate that both CSF and plasma provide a suitable medium for detection of ctDNA, and demonstrates the potential of liquid biopsy for identification of tumor-associated somatic mutations and replacing, or complementing current invasive and costly surgical biopsies.
Example 3: Assessment of treatment response using ctDNA quantification.
To investigate the clinical translation of liquid biopsy, we studied plasma obtained from patients diagnosed with DIPG who enrolled in an ongoing clinical trial (PNOC 003, NCT 2274987). Analysis of upfront and serial plasma samples probing for histone mutations (H3.3 and H3.1) indicated the following: i) histone mutations were successfully detected at diagnosis in 80% (n=16 of 20) of patients harboring known histone mutations determined by genomic analysis of pretreatment biopsies; ii) histone mutation was not detected at diagnosis in 20% of patients with histone mutations (n=4 of 20), hi) however, among these four patients, histone mutation was subsequently detected at post-radiation in two patients where serial samples were available (FIGS. 6D-E). MR image analysis of the two patients for whom ctDNA was absent at diagnosis, but present post-radiation indicated an increase in tumor volume following radiation (patient 12: 40.6mm3 to 62.4mm3, and patient 21: 28.6mm3 to 34.8mm3) (FIG. 6F), suggesting that changes in tumor architecture in response to radiation, or temporary disruption of the blood brain barrier (BBB) may contribute to the availability of ctDNA in plasma at different time points. Similarly, our analysis of serially collected CSF from a patient with H3 K27M DIPG at pre-treatment and at postmortem, revealed an increase in ctDNA abundance at the later stage of disease (FIGS. 10A- B).
To assess tumor response to treatment, longitudinal plasma samples obtained from enrolled patients (PNOC 003, NCT 2274987) at diagnosis, during treatment, and at tumor growth were studied (n=15). We found that in general, ctDNA abundances varied throughout the course of the treatment (see FIG.l 1). Importantly, patient- specific temporal analysis of changes in musculoaponeurotic fibrosarcoma oncogene (MAF) histone 3 mutation indicated a close association of ctDNA abundance with both clinical course of the disease, and tumor response as indicated by MR imaging (FIGS. 13A-D). Specifically, longitudinal ctDNA analysis of one patient showed a decrease in histone MAF post- radiation, stabilization during the first treatment course (panobinostat and mebendazole), a substantial increase at tumor growth, and a subsequent drop after patient underwent the second treatment course (FIG. 1 IB). To establish association of ctDNA MAF with tumor volume, a central MRI review was conducted to compare tumor volume (mm3) throughout the treatment course of patients enrolled in the clinical trial (PNOC 003, NCT 2274987) (FIG. 1 IB). In the nine patients who followed PNOC003 recommended therapy and had serial plasma samples collected throughout treatment, ctDNA abundance agreed with tumor volume reduction as assessed by MR imaging (FIG.11C).
To further compare ctDNA-based MAF with MR imaging, we performed statistical analysis of MAF (H3 ctDNA) at biopsy and post-radiation as well as tumor volume assessments using MR images. Our data showed that in 15 patients ctDNA MAF significantly decreased post- radiation when assessed by both liquid biopsy (p=0.004) and MR imaging (p=0.01) (Fig. 3d). Plasma ctDNA-MRI relationship was further compared by monitoring tumor response as assessed by MAF and MRI data at diagnosis and post-treatment (pre-cycle 3). Although both approaches indicated response, the MAF and tumor seize decrease were not significant (p=0.06 for decrease in ctDNA MAF and tumor volume measurements) (FIG. 1 IE).
We then assessed whether an increase in H3 K27M ctDNA accompanied tumor spread as determined by MRI, molecular, and/or histological studies. Tumor extension was determined by MRI obtained during the course of the treatment, or molecular and histopathological review of autopsied brain specimens. Tumor involvement beyond pons for patients enrolled in the clinical trial (PNOC 003, NCT 2274987) was determined by central MRI review at diagnosis, during therapy, or at tumor growth. We found that increased levels of CSF ctDNA corresponded to tumor spread; however, the small sample size for the availability of CSF for DIPGs with tumor dissemination precluded statistical significance. On the contrary, plasma collected at various times throughout disease (diagnosis, during treatment, or at tumor growth) was not predictive or indicative of tumor spread beyond pons (FIG. 1).
Outcomes for children diagnosed with MLG have not changed despite decades of clinical research. Recent advancements in genomic, epigenomic, and proteomic profiling, however, provide new opportunities for accelerating development of novel therapeutic interventions for childhood cancers. Collection of postmortem specimens resulted in the identification of oncohistones as driver mutations as well as other partner mutations, and newer clinical trials are building upon this knowledge to identify rational, biologically targeted treatment approaches.3' 4' 21 However, limited availability of biopsy in the pons and difficulty in accurately assessing patients' responses to therapy are current challenges in pediatric MLGs.
Liquid biopsy is an emerging tool for diagnosing, and measuring efficacy of treatment in adult cancer patients. While molecular profiling of tumors is a localized method, a liquid biopsy approach provides a systemic molecular overview. ctDNA has been used to determine patient's mutation profiles, as a biomarker for molecular-based disease monitoring, and recurrence in adult chronic lymphocytic leukemia, breast, and colon cancer.13'16'23 The only previously reported liquid biopsy approach for pediatric MLGs was established using Sanger sequencing.19 The disclosure herein for the detection and quantification of tumor-associated circulating DNA using ddPCR allows for rapid, more sensitive, far less costly and less invasive method for surveying tumor mutations, and represents a key advance particularly for tumors with limited tissue acquisition or prohibitive sampling at multiple time points.
While we report the detection of ctDNA in both CSF and plasma, the exact mechanism of tumor DNA release into the biofluid is not well understood. A survey of published manuscripts indicated that ctDNA is released from multiple sources, including living cells, apoptotic and/or necrotic tumor cells. We also found that CSF yielded a significantly higher amount of ctDNA compared to plasma. This is most likely due to the location of biofluids in relation to site of tumor, and challenges of overcoming the BBB for release of ctDNA into plasma.
Our lack of detection of ctDNA in plasma obtained at biopsy from four patients (H3 mutant by biopsy) may suggest high BBB integrity and thus lack of circulating tumor DNA. Subsequent analyses detected ctDNA in plasma drawn for two of these patients at post radiation, indicating the potential role of radiation therapy (RT) for temporarily disrupting BBB; hence, resulting in detection of ctDNA. Moreover, as the cellular turnover in a growing tumor increases, apoptotic and necrotic tumor cells are also responsible for the increased release of ctDNA into biofluids. As such, we expected to observe an initial spike in ctDNA as tumor cells die and release DNA into biofluids. However, our tested time points fell after the completion of RT when the tumor mass was reduced in most patients as assessed by MRI, corresponding to the expectant reduced number of ctDNA. An issue to be determined is whether inclusion of more time points during the course of RT, would detect an initial spike in ctDNA immediately after the first radiation treatment, followed by stabilization of ctDNA at maximal tumor response. Our study suggests that collection of multiple plasma draws at diagnosis and during treatment will be more informative as ctDNA may not be detectable in initial blood draws.
We have recently shown that DIPG tumor cells disseminate throughout the brain during the course of disease. Our ctDNA analysis in DIPG patients was indicative of tumor expansion beyond pons, where an increased amount of ctDNA in CSF was observed in patients who exhibited tumor spread. Studies of a larger cohort in clinical settings are required to assess the statistical significance of our finding. More importantly, our results indicate the unique strength of liquid biopsy for assessing the molecular landscape of MLGs, and potential for longitudinal assessment of tumor response to therapy, which is a new tool that is complementary to MR imaging. Similar to the clinical utility of ctDNA for monitoring response to therapy with respect to MRIs in adult GBMs, despite a small sample size, we found significant reduction in ctDNA following RT. Similar patterns in temporal changes of ctDNA and MRI assessments of tumor response indicated that our ddPCR analysis of plasma is highly translational, and offers a novel platform for assessing tumor response, regression and/or progression in MLGs. We provide a valuable tool that may be incorporated into future clinical trials for longitudinal assessment of treatment related molecular changes. Our liquid biopsy approach may also be expanded for facilitating diagnosis, longitudinal monitoring, and assessing recurrence in other childhood CNS cancers. In summary, we have shown that CSF and plasma ctDNA analysis of children with MLG is feasible, shows promise for detecting mutational load, provides an additional means for disease characterization, and, more importantly is a clinically relevant and sensitive method for assessing tumor response to treatment.
Figure imgf000030_0001
Table 2: Assessing specificity of a ddPCR platform by testing non-CNS malignant pediatric CSF and plasma.
Non-CNS
Diseased
Biofluid
Plasma Gene Mutation Average MAF type
Patient
ID
1252 Plasma H3F3A P.K27M 0.000%
1253 Plasma H3F3A P.K27M 0.000%
1255 Plasma H3F3A P.K27M 0.000%
1 140 Plasma HIST1H3B P.K27M 0.000%
1 141 Plasma HIST1H3B P.K27M 0.000%
1 142 Plasma HIST1H3B P.K27M 0.000%
1 143 Plasma HIST1H3B P.K27M 0.000%
1 144 Plasma HIST1H3B P.K27M 0.000%
1 145 Plasma HIST1H3B P.K27M 0.000%
1 146 Plasma HIST1H3B P.K27M 0.000%
1 147 Plasma HIST1H3B P.K27M 0.000%
1 148 Plasma HIST1H3B P.K27M 0.000%
942 CSF H3F3A P.K27M 0.000%
943 CSF H3F3A P.K27M 0.000%
945 CSF H3F3A P.K27M 0.000%
946 CSF H3F3A P.K27M 0.000%
947 CSF H3F3A P.K27M 0.000%
951 CSF H3F3A P.K27M 0.000%
955 CSF H3F3A P.K27M 0.001%
956 CSF H3F3A P.K27M 0.000%
853 CSF H3F3A P.K27M 0.000%
862 CSF H3F3A P.K27M 0.000%
856 CSF H3F3A P.K27M 0.000%
1 187 CSF H3F3A P.K27M 0.000%
1 195 CSF H3F3A P.K27M 0.000%
1 196 CSF H3F3A P.K27M 0.000%
1203 CSF H3F3A P.K27M 0.000%
1205 CSF H3F3A P.K27M 0.000%
943 CSF HIST1H3 P.K27M 0.000%
B
945 CSF HIST1H3 P.K27M 0.000%
B
946 CSF HIST1H3 P.K27M 0.000%
B Table 2: Assessing specificity of a ddPCR platform by testing non-CNS malignant pediatric CSF and plasma.
Non-CNS
Diseased
Biofluid
Plasma Gene Mutation Average MAF
type
Patient
ID
947 CSF HIST1H3 P.K27M 0.000%
B
951 CSF HIST1H3 P.K27M 0.000%
B
955 CSF HIST1H3 Ρ.Κ27Μ 0.001 %
B
956 CSF HIST1H3 Ρ.Κ27Μ 0.000%
B
853 CSF HIST1H3 Ρ.Κ27Μ 0.000%
B
862 CSF HIST1H3 Ρ.Κ27Μ 0.000%
B
856 CSF HIST1H3 Ρ.Κ27Μ 0.000%
B
1 187 CSF HIST1H3 Ρ.Κ27Μ 0.000%
B
Mutation allele frequencies (MAFs) for H3F3A p.K27M and HIST1H3B p.K27M in plasma and CSF collected from pediatric non-CNS diseased controls indicating the false positive rate of detection. Average MAF values for non- CNS diseased plasma analyzed for H3F3A p.K27M mutation represent technical triplicates, all other average MAF values for plasma and CSF analyzed for H3F3A and HIST1H3B p.K27M represent technical duplicates.
Figure imgf000033_0001
Figure imgf000034_0001
REFERENCES
I . Ostrom, Q.T., et al. American Brain Tumor Association Adolescent and Young Adult Primary Brain and Central Nervous System Tumors Diagnosed in the United States in 2008-2012. Neuro-oncology 18 Suppl 1, il-i50 (2016).
2. Castel, D., et al. Histone H3F3A and HIST1H3B K27M mutations define two subgroups of diffuse intrinsic pontine gliomas with different prognosis and phenotypes. Acta neuropathologica 130, 815-827 (2015).
3. Schwartzentruber, J., et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature 482, 226-231 (2012).
4. Wu, G., et al. Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas. Nature genetics 44, 251-253 (2012).
5. Louis, D.N., et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta neuropathologica 131, 803-820 (2016).
6. Plessier, A., et al. New in vivo avatars of diffuse intrinsic pontine gliomas (DIPG) from stereotactic biopsies performed at diagnosis. Oncotarget 8, 52543-52559 (2017).
7. Carai, A., et al. Robot- Assisted Stereotactic Biopsy of Diffuse Intrinsic Pontine GUoma: A Single-Center Experience. World neurosurgery 101, 584-588 (2017).
8. Hargrave, D., Chuang, N. & Bouffet, E. Conventional MRI cannot predict survival in childhood diffuse intrinsic pontine glioma. Journal of neuro-oncology 86, 313-319 (2008).
9. Laprie, A., et al. Longitudinal multivoxel MR spectroscopy study of pediatric diffuse brainstem gliomas treated with radiotherapy. International journal of radiation oncology, biology, physics 62, 20-31 (2005).
10. Riley, G.T., et al. Diffuse intrinsic pontine glioma: is MRI surveillance improved by region of interest volumetry? Pediatric radiology 45, 203-210 (2015).
I I. Chheda, Z.S., et al. Novel and shared neoantigen derived from histone 3 variant H3.3K27M mutation for glioma T cell therapy. The Journal of experimental medicine 215, 141- 157 (2018).
12. Ochs, K., et al. K27M-mutant histone-3 as a novel target for glioma
immunotherapy. Oncoimmunology 6, el 328340 (2017). 13. Tie, J., et al. Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage Π colon cancer. Science translational medicine, 346ra392 (2016).
14. Tsao, S.C., et al. Monitoring response to therapy in melanoma by quantifying circulating tumour DNA with droplet digital PCR for BRAF and NRAS mutations. Scientific reports 5, 11198 (2015).
15. Garcia-Murillas, I., et al. Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Science translational medicine 7, 302ral33 (2015).
16. Schiavon, G., et al. Analysis of ESR1 mutation in circulating tumor DNA demonstrates evolution during therapy for metastatic breast cancer. Science translational medicine 7, 313ral82 (2015).
17. De Mattos-Arruda, L., et al. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nature commumcations 6, 8839 (2015).
18. Diaz, L.A., Jr. & Bardelli, A. Liquid biopsies: genotyping circulating tumor DNA.
Journal of clinical oncology : official journal of the American Society of Clinical Oncology 32, 579-586 (2014).
19. Huang, T.Y., et al. Detection of Histone H3 mutations in cerebrospinal fluid- derived tumor DNA from children with diffuse midline glioma. Acta neuropathologica communications 5, 28 (2017).
20. Wang, W., Song, Z. & Zhang, Y. A Comparison of ddPCR and ARMS for detecting EGFR T790M status in ctDNA from advanced NSCLC patients with acquired EGFR-TKI resistance. Cancer medicine 6, 154-162 (2017).
21. Nikbakht, H., et al. Spatial and temporal homogeneity of driver mutations in diffuse intrinsic pontine glioma. Nature commumcations 7, 11185 (2016).
22. Saratsis, A.M., et al. Insights into pediatric diffuse intrinsic pontine glioma through proteomic analysis of cerebrospinal fluid. Neuro-oncology 14, 547-560 (2012).
23. Yeh, P., et al. Circulating tumour DNA reflects treatment response and clonal evolution in chronic lymphocytic leukaemia. Nature commumcations 8, 14756 (2017). 24. van der Vaart, M. & Pretorius, P.J. Circulating DNA. Its origin and fluctuation. Annals of the New York Academy of Sciences 1137, 18-26 (2008).
25. Jahr, S., et al. DNA fragments in the blood plasma of cancer patients:
quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer research 61, 1659-1665 (2001).
26. Stroun, M., Lyautey, J., Lederrey, C, Olson-Sand, A. & Anker, P. About the possible origin and mechanism of circulating DNA apoptosis and active DNA release. Clinica chimica acta; international journal of clinical chemistry 313, 139-142 (2001).

Claims

WHAT IS CLAIMED:
1. A method of determining the efficacy of a cancer therapy, said method comprising: a) administering a cancer therapy to a subject in need thereof;
b) obtaining a biological sample from the subject after administration of the cancer therapy to the subject; and
c) deteraiining the presence of a mutation in one or more genes selected from the group consisting of H3F3A, HIST1H3B, HIST1H3C, HIST2H3C, ACVRl, PPMID, PIK3R1, PIK3CA, IDH1, and BRAF in the biological sample,
wherein a decrease in the presence of the mutation(s) compared to the amount observed prior to administration of the cancer therapy indicates that the cancer therapy administered to the subject is effective.
2. The method of claim 1, wherein steps b) and c) are repeated at least once, wherein a decrease in the presence of the mutation(s) over time indicates that the cancer therapy administered to the subject is effective.
3. The method of claim 1, wherein said biological sample is a fluid.
4. The method of claim 3, wherein said biological fluid is blood, plasma, serum, CSF, or urine.
5. The method of claim 3, wherein said biological fluid is blood or plasma.
6. The method of claim 1 , wherein said cancer is a brain tumor.
7. The method of claim 6, wherein said cancer is a pediatric brain tumor.
8. The method of claim 6, wherein said brain tumor is a diffuse glioma or a diffuse intrinsic pontine glioma (DIPG).
9. The method of claim 1, wherein step c) comprises performing a method selected from the group consisting of quantitative polymerase chain reaction (qPCR), quantitative real-time polymerase chain reaction (qRTPCR), digital droplet PCR, (ddPCR), sequencing, northern blotting, and Southern blotting.
10. The method of claim 1, wherein step c) comprises determining the presence of a mutation in H3F3A.
11. The method of claim 10, wherein step c) comprises determining the presence of a K27M mutation in H3F3A.
12. The method of claim 11, wherein step c) further comprises determining the presence of a mutation in at least one other gene selected from the group consisting of HIST1H3B, HIST1H3C, HIST2H3C, ACVRl, PPM ID, PIK3R1, PIK3CA, IDH1, and BRAF.
13. The method of claim 11, wherein step c) further comprises determining the presence of a mutation in at least one other gene selected from the group consisting of HIST1H3B, ACVRl, PPM1D, PIK3R1, and PIK3CA.
14. The method of claim 11, wherein step c) further comprises determining the presence of a mutation in at least one other gene selected from the group consisting of HIST1H3B, ACVRl, PPMlD, and PIK3Rl.
15. A method of treating cancer in a subject in need thereof, said method comprising:
a) determining the presence of a mutation in one or more genes selected from the group consisting of H3F3A, HIST1H3B, HIST1H3C, HIST2H3C, ACVRl, PPM ID, PIK3R1, PIK3CA, IDH1, and BRAF in a biological sample, and
b) administering a cancer therapy to said subject if the mutation(s) is present.
16. The method of claim 15, wherein said biological sample is a fluid.
17. The method of claim 16, wherein said biological fluid is blood, plasma, serum, CSF, or urine.
18. The method of claim 16, wherein said biological fluid is blood or plasma.
19. The method of claim 15, wherein said cancer is a brain tumor.
20. The method of claim 19, wherein said cancer is a pediatric brain tumor.
21. The method of claim 19, wherein said brain tumor is a diffuse glioma or a diffuse intrinsic pontine glioma (DIPG).
22. The method of claim 15, wherein step a) comprises performing digital PCR or droplet digital PCR (ddPCR).
23. The method of claim 15, wherein step a) comprises determining the presence of a mutation in H3F3A.
24. The method of claim 22, wherein step a) comprises determining the presence of the K27M mutation in H3F3A.
25. The method of claim 23, wherein step a) further comprises determining the presence of a mutation in at least one other gene selected from the group consisting of HIST1H3B, HIST1H3C, HIST2H3C, ACVRl, PPM ID, PIK3R1, PIK3CA, IDH1, and BRAF.
26. The method of claim 23, wherein step a) further comprises determining the presence of a mutation in at least one other gene selected from the group consisting of HIST1H3B, ACVRl, PPM1D, PIK3R1, and PIK3CA.
27. The method of claim 23, wherein step a) further comprises determining the presence of a mutation in at least one other gene selected from the group consisting of HIST1H3B, ACVRl, PPMID, and PIK3R1.
28. A method of detecting a mutation in one or more genes selected from the group consisting of H3F3A, HIST1H3B, HIST1H3C, HIST2H3C, ACVRl, PPMID, PIK3R1, PIK3CA, IDHl, and BRAF in a blood or plasma sample from a human subject between 1 and 18 years of age, said method comprising performing digital PCR or droplet digital PCR (ddPCR) on the blood or plasma sample to determine the presence of a mutation in one or more genes selected from the group consisting of H3F3A, HIST1H3B, HIST1H3C, HIST2H3C, ACVRl, PPMID, PIK3R1, PIK3CA, IDHl, and BRAF in a biological sample.
29. The method of claim 28, wherein the subject has cancer.
30. The method of claim 29, wherein the cancer is a brain tumor.
31. The method of claim 30, wherein said brain tumor is a diffuse glioma or a diffuse intrinsic pontine glioma (DIPG).
32. The method of claim 28, wherein the one or more genes comprise H3F3A.
33. The method of claim 32, wherein the mutation is a K27M mutation in H3F3A.
34. The method of claim 33, further comprising determining the presence of a mutation in at least one other gene selected from the group consisting of HIST1H3B, HIST1H3C, HIST2H3C,
ACVRl, PPMID, PIK3R1, PIK3CA, IDHl, and BRAF.
35. The method of claim 33, further comprising determining the presence of a mutation in at least one other gene selected from the group consisting of HIST1H3B, ACVRl, PPMID, PIK3R1, and PIK3CA.
36. The method of claim 33, wherein step c) further comprises determining the presence of a mutation in at least one other gene selected from the group consisting of HIST1H3B, ACVRl, PPMW, and PIK3Rl.
37. An oligonucleotide probe comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 19-36, wherein the probe further comprises a label.
38. A composition comprising at least one oligonucleotide probe of claim 37.
PCT/US2018/051150 2017-09-15 2018-09-14 Methods for detecting cancer biomarkers WO2019055829A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762559049P 2017-09-15 2017-09-15
US62/559,049 2017-09-15

Publications (1)

Publication Number Publication Date
WO2019055829A1 true WO2019055829A1 (en) 2019-03-21

Family

ID=63788032

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/051150 WO2019055829A1 (en) 2017-09-15 2018-09-14 Methods for detecting cancer biomarkers

Country Status (1)

Country Link
WO (1) WO2019055829A1 (en)

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US6306597B1 (en) 1995-04-17 2001-10-23 Lynx Therapeutics, Inc. DNA sequencing by parallel oligonucleotide extensions
US6797470B2 (en) 1996-05-29 2004-09-28 Cornell Research Foundation, Inc. Detection of nucleic acid sequence differences using coupled ligase detection and polymerase chain reactions
US7011944B2 (en) 1996-01-24 2006-03-14 Third Wave Technologies, Inc. Invasive cleavage of nucleic acids
US7037687B2 (en) 1998-05-01 2006-05-02 Arizona Board Of Regents Method of determining the nucleotide sequence of oligonucleotides and DNA molecules
US7083917B2 (en) 1996-02-09 2006-08-01 Cornell Research Foundation, Inc. Detection of nucleic acid sequence differences using the ligase detection reaction with addressable arrays
US7169560B2 (en) 2003-11-12 2007-01-30 Helicos Biosciences Corporation Short cycle methods for sequencing polynucleotides
US7232656B2 (en) 1998-07-30 2007-06-19 Solexa Ltd. Arrayed biomolecules and their use in sequencing
US7598035B2 (en) 1998-02-23 2009-10-06 Solexa, Inc. Method and compositions for ordering restriction fragments
US7645596B2 (en) 1998-05-01 2010-01-12 Arizona Board Of Regents Method of determining the nucleotide sequence of oligonucleotides and DNA molecules
US7769400B2 (en) 2004-11-22 2010-08-03 Seven Networks International Oy Connectivity function for forwarding e-mail
CN104031992B (en) * 2014-05-27 2016-03-09 武汉海吉力生物科技有限公司 Mankind B-raf gene V600 mutation detection kit
US9984201B2 (en) 2015-01-18 2018-05-29 Youhealth Biotech, Limited Method and system for determining cancer status

Patent Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683202B1 (en) 1985-03-28 1990-11-27 Cetus Corp
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US6306597B1 (en) 1995-04-17 2001-10-23 Lynx Therapeutics, Inc. DNA sequencing by parallel oligonucleotide extensions
US7011944B2 (en) 1996-01-24 2006-03-14 Third Wave Technologies, Inc. Invasive cleavage of nucleic acids
US7083917B2 (en) 1996-02-09 2006-08-01 Cornell Research Foundation, Inc. Detection of nucleic acid sequence differences using the ligase detection reaction with addressable arrays
US7364858B2 (en) 1996-05-29 2008-04-29 Cornell Research Foundation, Inc. Detection of nucleic acid sequence differences using coupled ligase detection and polymerase chain reactions
US6797470B2 (en) 1996-05-29 2004-09-28 Cornell Research Foundation, Inc. Detection of nucleic acid sequence differences using coupled ligase detection and polymerase chain reactions
US7166434B2 (en) 1996-05-29 2007-01-23 Cornell Research Foundation, Inc. Detection of nucleic acid sequence differences using coupled ligase detection and polymerase chain reactions
US7429453B2 (en) 1996-05-29 2008-09-30 Cornell Research Foundation, Inc. Detection of nucleic acid sequence differences using coupled ligase detection and polymerase chain reactions
US7320865B2 (en) 1996-05-29 2008-01-22 Cornell Research Foundation, Inc. Detection of nucleic acid sequence differences using coupled ligase detection and polymerase chain reactions
US7332285B2 (en) 1996-05-29 2008-02-19 Cornell Research Foundation, Inc. Detection of nucleic acid sequence differences using coupled ligase detection and polymerase chain reactions
US7598035B2 (en) 1998-02-23 2009-10-06 Solexa, Inc. Method and compositions for ordering restriction fragments
US7645596B2 (en) 1998-05-01 2010-01-12 Arizona Board Of Regents Method of determining the nucleotide sequence of oligonucleotides and DNA molecules
US7037687B2 (en) 1998-05-01 2006-05-02 Arizona Board Of Regents Method of determining the nucleotide sequence of oligonucleotides and DNA molecules
US7232656B2 (en) 1998-07-30 2007-06-19 Solexa Ltd. Arrayed biomolecules and their use in sequencing
US7169560B2 (en) 2003-11-12 2007-01-30 Helicos Biosciences Corporation Short cycle methods for sequencing polynucleotides
US7769400B2 (en) 2004-11-22 2010-08-03 Seven Networks International Oy Connectivity function for forwarding e-mail
CN104031992B (en) * 2014-05-27 2016-03-09 武汉海吉力生物科技有限公司 Mankind B-raf gene V600 mutation detection kit
US9984201B2 (en) 2015-01-18 2018-05-29 Youhealth Biotech, Limited Method and system for determining cancer status

Non-Patent Citations (57)

* Cited by examiner, † Cited by third party
Title
BAKER, M., NAT METH., vol. 9, 2012, pages 541 - 544
BENDER ET AL., CANCER CELL, vol. 24, no. 5, 2013, pages 660 - 72
BETTEGOWDA ET AL., SCI TRANSL. MED., vol. 6, no. 224, 2014, pages 224ra24
BIBKOVA ET AL., GENOME RES, vol. 16, 2006, pages 383 - 393
CARAI, A. ET AL.: "Robot-Assisted Stereotactic Biopsy of Diffuse Intrinsic Pontine Glioma: A Single-Center Experience", WORLD NEUROSURGERY, vol. 101, 2017, pages 584 - 588, XP085025884, DOI: doi:10.1016/j.wneu.2017.02.088
CASTEL, D. ET AL.: "Histone H3F3A and HIST1H3B K27M mutations define two subgroups of diffuse intrinsic pontine gliomas with different prognosis and phenotypes", ACTA NEUROPATHOLOGICA, vol. 130, 2015, pages 815 - 827
CHHEDA, Z.S. ET AL.: "Novel and shared neoantigen derived from histone 3 variant H3.3K27M mutation for glioma T cell therapy", THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 215, 2018, pages 141 - 157
DE MATTOS-ARRUDA ET AL., NAT. COMMUN., vol. 6, 2015, pages 8839
DE MATTOS-ARRUDA, L. ET AL.: "Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma", NATURE COMMUNICATIONS, vol. 6, 2015, pages 8839
DIAZ ET AL., J. CLIN. ONCOL., vol. 32, no. 6, 2014, pages 579 - 86
DIAZ, L.A., JR.; BARDELLI, A.: "Liquid biopsies: genotyping circulating tumor DNA. Journal of clinical oncology : official journal of the", AMERICAN SOCIETY OF CLINICAL ONCOLOGY, vol. 32, 2014, pages 579 - 586
E.W. MARTIN: "Remington's Pharmaceutical Sciences", MACK PUBLISHING CO.
ESHINI PANDITHARATNA ET AL: "DIPG-39. LIQUID BIOPSY FOR MONITORING OF TUMOR RESPONSE IN CHILDREN WITH MIDLINE GLIOMAS", NEURO-ONCOLOGY, 1 June 2017 (2017-06-01), pages iv13 - iv14, XP055531534, Retrieved from the Internet <URL:https://academic.oup.com/neuro-oncology/article-pdf/19/suppl_4/iv13/17642768/nox083.054.pdf> [retrieved on 20181207] *
GARCIA-MURILLAS ET AL., SCI TRANSL MED., vol. 7, no. 302, 2015, pages 302ra133
GARCIA-MURILLAS, I. ET AL.: "Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer", SCIENCE TRANSLATIONAL MEDICINE, vol. 7, 2015, pages 302ra133, XP055440860, DOI: doi:10.1126/scitranslmed.aab0021
GENNARO, A. R.: "Remington: The Science and Practice of Pharmacy", LIPPINCOTT, WILLIAMS AND WILKINS
HAMID NIKBAKHT ET AL: "Spatial and temporal homogeneity of driver mutations in diffuse intrinsic pontine glioma", NATURE COMMUNICATIONS, vol. 7, no. 1, 6 April 2016 (2016-04-06), XP055531561, DOI: 10.1038/ncomms11185 *
HARGRAVE, D.; CHUANG, N.; BOUFFET, E.: "Conventional MRI cannot predict survival in childhood diffuse intrinsic pontine glioma", JOURNAL OF NEURO-ONCOLOGY, vol. 86, 2008, pages 313 - 319, XP019578969
HARRIS ET AL., SCIENCE, vol. 320, 2008, pages 106 - 109
HINSON ET AL., ANAL. CHEM., vol. 83, 2011, pages 8604 - 8610
HUANG, T.Y. ET AL.: "Detection of Histone H3 mutations in cerebrospinal fluid-derived tumor DNA from children with diffuse midline glioma", ACTA NEUROPATHOLOGICA COMMUNICATIONS, vol. 5, 2017, pages 28
INNIS ET AL,: "PCR Protocols, A Guide to Methods and Application", 1990, ACADEMIC PRESS, INC.
JAHR, S. ET AL.: "DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells", CANCER RESEARCH, vol. 61, 2001, pages 1659 - 1665, XP002494286
KAUPPINEN ET AL., DRUG DISCOV. TODAY TECH., vol. 2, 2005, pages 287 - 290
KIBBE, ET AL.: "Handbook of Pharmaceutical Excipients", AMERICAN PHARMACEUTICAL ASSOCIATION
LAPRIE, A. ET AL.: "Longitudinal multivoxel MR spectroscopy study of pediatric diffuse brainstem gliomas treated with radiotherapy", INTERNATIONAL JOURNAL OF RADIATION ONCOLOGY, BIOLOGY, PHYSICS, vol. 62, 2005, pages 20 - 31, XP025262868, DOI: doi:10.1016/j.ijrobp.2004.09.027
LETICIA DE MATTOS-ARRUDA ET AL: "Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma", NATURE COMMUNICATIONS, vol. 6, no. 1, 10 November 2015 (2015-11-10), XP055531832, DOI: 10.1038/ncomms9839 *
LEWIS ET AL., SCIENCE, vol. 340, no. 6134, 2013, pages 857 - 61
LIBERMAN, ET AL.: "Pharmaceutical Dosage Forms", MARCEL DECKER
LOUIS, D.N. ET AL.: "The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary", ACTA NEUROPATHOLOGICA, vol. 131, 2016, pages 803 - 820, XP035878676, DOI: doi:10.1007/s00401-016-1545-1
MARGULIES ET AL., NATURE, vol. 437, 2005, pages 376 - 380
MURTAZA ET AL., NATURE, vol. 497, no. 7447, 2013, pages 108 - 12
NIKBAKHT ET AL., NAT. COMMUN., vol. 7, 2016, pages 11185
NIKBAKHT, H. ET AL.: "Spatial and temporal homogeneity of driver mutations in diffuse intrinsic pontine glioma", NATURE COMMUNICATIONS, vol. 7, 2016, pages 11185
OCHS, K. ET AL.: "K27M-mutant histone-3 as a novel target for glioma immunotherapy", ONCOIMMUNOLOGY, vol. 6, 2017, pages e1328340
ORUM ET AL., LETTERS PEPTIDE SCI., vol. 10, 2004, pages 325 - 334
OSTROM, Q.T. ET AL.: "American Brain Tumor Association Adolescent and Young Adult Primary Brain and Central Nervous System Tumors Diagnosed in the United States in 2008-2012", NEURO-ONCOLOGY, vol. 18, no. 1, 2016, pages il - i50
PINHEIRO ET AL., ANAL. CHEM., vol. 84, 2012, pages 1003 - 1011
PLESSIER, A. ET AL.: "New in vivo avatars of diffuse intrinsic pontine gliomas (DIPG) from stereotactic biopsies performed at diagnosis", ONCOTARGET, vol. 8, 2017, pages 52543 - 52559
RILEY, G.T. ET AL.: "Diffuse intrinsic pontine glioma: is MRI surveillance improved by region of interest volumetry?", PEDIATRIC RADIOLOGY, vol. 45, 2015, pages 203 - 210, XP035438485, DOI: doi:10.1007/s00247-014-3134-6
SARATSIS, A.M. ET AL.: "Insights into pediatric diffuse intrinsic pontine glioma through proteomic analysis of cerebrospinal fluid", NEURO-ONCOLOGY, vol. 14, 2012, pages 547 - 560
SCHIAVON, G. ET AL.: "Analysis of ESR1 mutation in circulating tumor DNA demonstrates evolution during therapy for metastatic breast cancer", SCIENCE TRANSLATIONAL MEDICINE, vol. 7, 2015, pages 313ra182, XP055504206, DOI: doi:10.1126/scitranslmed.aac7551
SCHIOVAN ET AL., SCI. TRANSL. MED., vol. 7, no. 313, 2015, pages 313ral82
SCHWARTZENTRUBER, J. ET AL.: "Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma", NATURE, vol. 482, 2012, pages 226 - 231
SCOTT RYALL ET AL: "Targeted detection of genetic alterations reveal the prognostic impact of H3K27M and MAPK pathway aberrations in paediatric thalamic glioma", ACTA NEUROPATHOLOGICA COMMUNICATIONS, vol. 4, no. 1, 31 August 2016 (2016-08-31), XP055531268, DOI: 10.1186/s40478-016-0353-0 *
SONI; MELLER, CLIN. CHEM., vol. 53, 2007, pages 1996 - 2001
STROUN, M.; LYAUTEY, J.; LEDERREY, C.; OLSON-SAND, A.; ANKER, P.: "About the possible origin and mechanism of circulating DNA apoptosis and active DNA release", CLINICA CHIMICA ACTA; INTERNATIONAL JOURNAL OF CLINICAL CHEMISTRY, vol. 313, 2001, pages 139 - 142
TIE ET AL., SCI. TRANSL. MED., vol. 8, no. 346, 2016, pages 346ra92
TIE, J. ET AL.: "Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage Π colon cancer", SCIENCE TRANSLATIONAL MEDICINE, 2016, pages 346ra392
TINAY HUANG ET AL: "Detection of Histone H3 mutations in cerebrospinal fluid-derived tumor DNA from children with diffuse midline glioma", ACTA NEUROPATHOLOGICA COMMUNICATIONS, BIOMED CENTRAL LTD, LONDON, UK, vol. 5, no. 1, 17 April 2017 (2017-04-17), pages 1 - 12, XP021244168, DOI: 10.1186/S40478-017-0436-6 *
TSAO ET AL., SCI. REP., vol. 5, 2015, pages 11198
TSAO, S.C. ET AL.: "Monitoring response to therapy in melanoma by quantifying circulating tumour DNA with droplet digital PCR for BRAF and NRAS mutations", SCIENTIFIC REPORTS, vol. 5, 2015, pages 11198
VAN DER VAART, M.; PRETORIUS, P.J.: "Circulating DNA. Its origin and fluctuation", ANNALS OF THE NEW YORK ACADEMY OF SCIENCES, vol. 1137, 2008, pages 18 - 26
WANG, W.; SONG, Z.; ZHANG, Y.: "A Comparison of ddPCR and ARMS for detecting EGFR T790M status in ctDNA from advanced NSCLC patients with acquired EGFR-TKI resistance", CANCER MEDICINE, vol. 6, 2017, pages 154 - 162
WU, G. ET AL.: "Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas", NATURE GENETICS, vol. 44, 2012, pages 251 - 253, XP055151998, DOI: doi:10.1038/ng.1102
YEH, P. ET AL.: "Circulating tumour DNA reflects treatment response and clonal evolution in chronic lymphocytic leukaemia", NATURE COMMUNICATIONS, vol. 8, 2017, pages 14756
ZOU ET AL., ASSOCIATION OF CLINICAL CHEMISTRY (AACC) POSTER PRESENTATION ON, 28 July 2010 (2010-07-28)

Similar Documents

Publication Publication Date Title
US11512349B2 (en) Methods for detecting disease using analysis of RNA
US11111541B2 (en) Diagnostic MiRNA markers for Parkinson&#39;s disease
Sefrioui et al. Clinical value of chip-based digital-PCR platform for the detection of circulating DNA in metastatic colorectal cancer
US20180282820A1 (en) Monitoring treatment or progression of myeloma
US10457988B2 (en) MiRNAs as diagnostic markers
EP3122905B1 (en) Circulating micrornas as biomarkers for endometriosis
US20190226034A1 (en) Proteomics analysis and discovery through dna and rna sequencing, systems and methods
US20230399702A1 (en) Method for screening a subject for cancer
US20160040253A1 (en) Method for manufacturing gastric cancer prognosis prediction model
AU2019372440A1 (en) Methods to diagnose and treat cancer using non-human nucleic acids
US20170130269A1 (en) Diagnosis of neuromyelitis optica vs. multiple sclerosis using mirna biomarkers
Zheng et al. Clinical applications of liquid biopsies for early lung cancer detection
KR20190077309A (en) Diagnosis and treatment of esophageal cancer
US10161004B2 (en) Diagnostic miRNA profiles in multiple sclerosis
AU2024203201A1 (en) Multimodal analysis of circulating tumor nucleic acid molecules
US20210375391A1 (en) Detection of microsatellite instability
KR102096498B1 (en) MicroRNA-4732-5p for diagnosing or predicting recurrence of colorectal cancer and use thereof
Tang et al. Maximum allele frequency observed in plasma: A potential indicator of liquid biopsy sensitivity
TW201514311A (en) Method for determining the prognosis of pancreatic cancer
WO2019055829A1 (en) Methods for detecting cancer biomarkers
JP2023507796A (en) Multiplex assay using different fragment sizes to identify cancer-specific cell-free DNA
KR102096499B1 (en) MicroRNA-3960 for diagnosing or predicting recurrence of colorectal cancer and use thereof
CN102864236B (en) Molecular marker for detecting susceptibility of ovarian cancer
Badalamenti et al. Cell-free/circulating tumor DNA profiling: from next-generation sequencing-based to digital polymerase chain reaction-based methods
WO2023224488A1 (en) Dna repair signature and prediction of response following cancer therapy

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18782854

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18782854

Country of ref document: EP

Kind code of ref document: A1